Anda di halaman 1dari 13

Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432

Seminar article
Targeted therapies for kidney cancer in urologic practice
Naomi B. Haas, M.D.a, Robert G. Uzzo, M.D.b,*
a
Department of Medical Oncology, Fox Chase Cancer Center, Temple University School of Medicine, Philadelphia, PA 19111, USA
b
Department of Urologic Oncology, Fox Chase Cancer Center, Temple University School of Medicine, Philadelphia, PA 19111, USA

Abstract
Renal cell carcinoma (RCC) is the most lethal of all genitourinary malignancies with nearly half of all patients presenting with locally
advanced or metastatic disease. Systemic treatments such as chemo- or immunotherapy have historically been associated with overall
response rates of 5–15% with very few durable responses. The basis of newly approved, more effective targeted therapies for metastatic
RCC are based on a fundamental knowledge of the molecular mechanisms that give rise to RCC. We review the clinical data for targeted
therapies in RCC and discuss the pertinent biology, side effects, and targets important to the practicing clinician. © 2007 Elsevier Inc. All
rights reserved.

Keywords: Kidney; Renal; Cancer; Targeted therapy; Angiogenesis

Introduction patients with early, localized disease, radical nephrectomy


is associated with a 5-year cancer-specific survival (CSS) as
Kidney cancer accounts for 3% of all solid tumors with high as 97% for pT1a lesions and 87% for pT1b tumors,
an estimated 36,160 new cases and 12,660 deaths per year, while nephron-sparing surgery (NSS) is associated with 5-
making renal cell carcinoma (RCC) the most lethal of all and 10-year CSS of 96% and 90%, respectively, for tumors
genitourinary neoplasms [1]. Like most malignancies, RCC ⱕ4 cm [9]. Early data regarding laparoscopic partial ne-
is a heterogeneous disease. This heterogeneity is reflected in phrectomy is similarly favorable, albeit with more limited
its presentation, pathology, molecular biology, and clinical follow-up [10]. Minimally invasive ablative technologies
course. Although most patients are completely asymptom- are emerging as potential treatment options for localized
atic, many may exhibit a variety of symptoms at presenta- RCC with excellent initial outcomes [11].
tion, including any of the classic triad of flank pain, hema- Unfortunately, 20% of patients have either locally ad-
turia, and a palpable abdominal mass. Other nonspecific vanced or node positive (N⫹) RCC while another 22% have
presenting symptoms include fatigue, weight loss, or ane- metastatic RCC (mRCC) at presentation [1]. Unlike the
mia [2]. The widespread use of body imaging modalities has outcomes in early localized disease, survival rates for N⫹
led to an increased detection of incidental renal masses over the patients are poor and patients with mRCC are rarely cured
last two decades [3]. Pathologic heterogeneity is illustrated by despite aggressive multimodal therapy. Classic cytotoxic
several distinct histological subtypes, which exhibit varying chemotherapy has repeatedly been shown to have little
degrees of biological aggressiveness [4] and by variations in effect in RCC [12,13]. Cytoreductive nephrectomy with
tumor grade [5]. Emerging data from molecular analyses indi- systemic immunotherapy is associated with few cures and
cate that RCCs express a variety of molecular tumor markers poor CSS outcomes with median survivals of 12 to 24
and unique patterns of gene expression [6,7]. Clinically, the months [14]. Moreover, only 5% to 20% of patients with
disease behaves quite heterogeneously, with courses ranging mRCC respond to biological agents such as interferon
from indolent [8] to highly aggressive. and/or interleukin [15,16]. A recent meta-analysis of 53
For most cases of RCC, surgical monotherapy or as part published randomized clinical trials that stratified 6,117
of a multimodal approach remains the standard of care. In patients with advanced RCC to an immunotherapeutic agent
in at least one arm demonstrated an overall chance of partial
* Corresponding author. Tel.: ⫹1-215-728-3501; fax: ⫹1-215-214-
or complete response to immunotherapies of only 12.9%
1734. compared with 4.3% in the placebo arm with a median
E-mail address: Robert.Uzzo@fccc.edu (R.G. Uzzo). survival of 13 months [16].

1078-1439/07/$ – see front matter © 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.urolonc.2007.05.009
N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432 421

Despite this rather poor track record in the management A small number of established genetic mechanisms are
of advanced RCC, recent advances in our understanding of responsible for the majority of known mutations in kidney
the molecular origins and pathways of RCC have led to the cancer related tumor suppressors or oncogenes. These in-
development of more effective targeted therapies. Here we clude loss of heterozygosity (LOH), homozygous deletion,
review the molecular pathways that define the pertinent rearrangement, point mutation, and promoter hypermethyl-
therapeutic targets in RCC and the clinical data for these ation [21]. Loss of heterozygosity (LOH) involves the loss
new and promising approaches. of specific chromosomal regions that contain known or
presumptive tumor suppressor genes. LOH may be achieved
through a deletion, gene conversion, mitotic recombination,
Genetics, pathways, and targets in RCC translocation, chromosome breakage and loss, chromosomal
fusion or telomeric end-to-end fusions, or whole chromo-
Our understanding of the genetics and molecular pathways some loss. LOH is a useful diagnostic marker because of its
in RCC is perhaps more advanced than for any other solid high level of specificity for transformed cells. Point muta-
malignancy. Unraveling these pathways has been possible only tions occur at the level of individual nucleotides where
through a series of sentinel observations made over the course substitutions, insertions, and deletions may result in frame-
of the last three decades. Some knowledge of the basic biology shift, missense, and nonsense mRNA transcripts, and ulti-
of RCC is essential for clinicians who manage this disease, mately truncated or non-functional proteins. Promoter hy-
particularly in this new era of targeted therapies. permethylation is another point of genetic control and
counter-control [22]. In most somatic cells, the CpG dinu-
Basic molecular biology cleotide “islands” in the controlling or promoter region of a
gene are normally protected from methylation. Although
The study of hereditary forms of RCC has been essential not a distinct genetic event, promoter hypermethylation is
in understanding the molecular origins of this disease. Iden- an epigenetic event capable of silencing expression. One or
tification of affected family pedigrees has led to the discov- more of these mechanisms may be responsible for an inher-
ery of germline mutations in genes that predispose individ- ited (germline) or acquired (somatic) mutation in tumor
uals to tumor development. To date, several inherited forms suppressor or oncogenes; however, LOH and hypermethyl-
of RCC have been described, including von Hippel Lindau ation are not typically associated with oncogenes.
(VHL), hereditary papillary renal carcinoma (HPRC), the
Birt-Hogg Dubet syndrome (BHD), and hereditary leiomy- Genetics of histologic subtypes of RCC
oma renal cell carcinoma (HLRCC) [17]. The specific mu-
tations involved are distinct and may involve a loss of Clear cell carcinoma
function event (tumor suppressor) such as in VHL, or a gain The VHL gene was initially mapped to the short arm of
of function event (oncogene) as in HPRC. Most tumor chromosome 3 (3p) by Seizinger et al. in 1988 following
suppressor genes (including VHL) follow Knudson’s “two reports of 3p deletions in sporadic RCC [23]. Genetic link-
hit” theory of tumorigenesis, which postulates that certain age and in situ hybridization studies subsequently mapped it
genes encode proteins responsible for the negative regula- to the 3p25 locus with LOH observed in greater than 95% of
tion of cell growth [18]. Mutations causing loss of function cases of both early- and late-stage sporadic clear cell car-
of these proteins result in uninhibited cell growth and ma- cinoma [24]. Isolation of the VHL gene was ultimately
lignant transformation. Mutation of a tumor suppressor gene accomplished using a positional cloning strategy. It is now
on one chromosome is insufficient to induce tumorigenesis; known that differences in phenotype between patients with
however, if both alleles are affected, tumor formation will VHL are due to variable mutations in a single gene rather
ensue. Because patients with familial cancers such as VHL than multiple genes. Therefore, allelic heterogeneity rather
inherit a germline mutation, they only require one “hit” to than locus heterogeneity is responsible for differences in the
inactivate the remaining tumor suppressor gene. However, clinical manifestations of VHL [25].
patients with sporadic tumors require two “hits,” which The VHL gene is small and is composed of 3 exons with
occur less frequently [19]. For this reason, patients with 852 coding nucleotides [26]. Recent studies have demon-
familial cancers typically present at an earlier age with strated somatic mutations of the VHL gene in approxi-
multifocal and/or bilateral disease. mately 50% to 75% of cases of sporadic clear cell renal
In contrast to mutations of tumor suppressors, mutated carcinoma [27]. Most sporadic clear cell cancers have either
proto-oncogenes (oncogenes) are responsible for gain of point mutations of the VHL gene, hypermethylation of the
function processes leading to increased cellular growth, promoter region, or rearrangement of the gene. Notably,
differentiation or proliferation through the accumulation of genetic “hotspots” exist where individual mutations cluster,
normal signaling proteins, or creation of a mutant activating principally at the 3= end of exon 1 and the 5= end of exon 3;
protein as in the case of the met gene in HPRC [20]. Unlike however, specific mutations at nucleotides 712 and 713
tumor suppressor genes, oncogenes may only require a (codon 238) may account for nearly half of all mutations in
single mutating event to promote malignant transformation. patients with VHL associated pheochromocytoma [26].
422 N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432

Table 1
Molecular targets of current targeted therapies
Target Sunitinib Sorafenib AG013736 GW786034 ABT-869 PTK-787 Bevacizumab Temsirolimus/
[34] [35] [36] [37,38] everolimus

Vascular endothelial growth N N N N N N Y N


factor (VEGF)
VEGFR1 (Flt-1) Y N Y Y Y Y N N
VEGFR2 N Y Y Y Y Y N N
(Flk-1/KDR)
VEGFR3 (FLT-4) Y Y Y Y N N
Platelet derived growth factor N N Y N N N
receptor ␣
PDGFR-␤ Y Y Y Y Y N N N
c-kit Y Y Y Y Y N N N
FLT-3 Y Y Y U Y N N N
Receptor stem cell factor (SCF) Y N U U N N N
RET Y N Y N U N N N
FAK (focal adhesion kinase) N N N N U N N N
Basic fibroblast growth factor Y Y Y Y Y N N N
(b-FGF)
B-raf kinase N Y N N N N N N
c-raf kinase N Y N N N N N N
Mammalian target of rapamycin N N N N N N N Y
(MTOR)

Y ⫽ inhibits target; N ⫽ no inhibition; U ⫽ unknown.

Other genetic alterations in clear cell RCC include frequent [32]. Therefore, the finding of LOH on numerous chromo-
LOH of chromosomes 8p, 14q, and 9p (p16) among others somes supports the diagnosis of chromophobe carcinoma.
[28], suggesting other unknown tumor suppressors may be Collecting duct carcinomas or Bellini tumors are un-
important in renal cell tumorigenesis. common and clinically very aggressive cancers [33]. The
cytogenetics of three collecting duct cancers have been
Papillary RCC characterized [32]. Each was consistently monosomic for
The most common genetic events associated with pap- chromosomes 1, 6, 14, 15, and 22. Trisomy 17, tri- or
illary renal cancer are trisomy of chromosomes 7 and 17 tetrasomy 7, or deletion of chromosome 3p were not ob-
and loss of Y [28]. Linkage analyses in families with served by cytogenetic analysis, thus differentiating these
HPRC have localized the site for the HPRC gene to 7q31. tumors from papillary and clear cell carcinoma, respec-
The HPRC gene has been identified as the c-met proto- tively. Monosomy of chromosome 1 appears to be a com-
oncogene. Mutations of the met oncogene are perhaps mon finding in collecting duct carcinoma [33].
best studied in familial papillary renal cancer with occa-
sional mutations found in apparently sporadic cases. Ac- VEGF and pertinent pathways in RCC
tivating missense mutations in the important intracellular
signaling tyrosine kinase domain of the met gene may be Transcription and translation of mutated tumor sup-
responsible for the gain of function events in papillary pressor genes or oncogenes ultimately lead to malignant
renal carcinoma [29]. transformation through aberrant protein expression.
While it was once believed that there was a precise
Chromophobe and collecting duct carcinoma relationship between a gene and its protein product, it is
Chromophobe carcinoma is associated with multiple now known that through a process known as alternative
chromosomal losses, most commonly monosomy of multi- splicing, there is significant redundancy in gene expres-
ple chromosomes, including 1, 2, 6, 10, 13, 17, and 21. sion such that one gene may be responsible for any
Frequent LOH of chromosomal arms 3p, 5q, 17p, and 17q number of protein products and their isoforms. This has
has been identified in chromophobe tumors as has loss of hampered the study of cellular proteins responsible for
chromosomes 1 (100% of cases), 2 (95% of cases), 6, 10, the malignant transformation of renal epithelium. None-
13, 17, and 21 [30]. Van den Berg et al. found the more theless, a number of known proteins are believed to be
frequent aberrations to be polysomy 7 and trisomies of 12, under the control of mutated genes implicated in the
16, 18, and 19, structural abnormalities on 11q and telo- development of RCC. These proteins represent rationale
meric association involving chromosome 7 [31]. Genetic targets for new therapies in RCC (Table 1).
deletions on chromosome 3 have been reported in 25% of Transformation may result from abnormal expression of
chromophobe tumors, but VHL gene mutations are absent normal cellular proteins, normal expression of abnormal
N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432 423

proteins, or both. Ordinarily, the VHL gene product (pVHL) demonstrated 40% partial response as assessed strictly with
forms a multimer (VEC) with Elongin B and C, Cullin 2, RECIST [49] criteria and a median duration of response of
and Rbx1, which binds to the ␣ subunits of HIF1, a tran- 8.7 months. The second Phase II RCC trial enrolled a
scription factor that allows cells to grow and survive under similar population of 106 patients with identical dosing and
hypoxic conditions. Under normal oxygen conditions, hy- demonstrated a response rate of 44% with one complete
droxylation of HIF allows subsequent binding to VEC, response by RECIST, and a median time to progression of
which marks it for ubiquitination and degradation. Under 8.1 months. A randomized Phase III trial of interferon-␣
low oxygen states or in the presence of mutated pVHL, (IFN-␣) (9 million units TIW) vs. the same schedule of
HIF1␣ cannot bind VEC, which is then free to translocate sunitinib in 690 patients with first-line metastatic clear cell
into the nucleus and up-regulate production of its targeted RCC (presented at ASCO in 2006) showed a median pro-
genes, including VEGF and other hypoxia inducible pro- gression-free survival (PFS) of 47.3 weeks for sunitinib vs.
teins such as TGF-␣, PDGF-␤, CAIX (G250), and the 24.9 weeks for IFN-␣. The objective response rate (ORR)
glucose transporter Glut-1 [39,40]. by third-party independent review was 24.8% for sunitinib
Many of the receptors for HIF inducible factors exhibit vs. 4.9% for IFN-␣. Thus, this trial demonstrated a signif-
tyrosine binding activity, which, upon ligand binding, acti- icant improvement in PFS and ORR for sunitinib over
vate downstream signaling pathways such as the Raf/MEK/ IFN-␣ in first-line treatment of patients with metastatic
ERK pathway. This process of signal transduction through RCC and is now indicated as first-line treatment for meta-
the receptor forms a link between extracellular signals on static RCC [50]. Additional schedules are being explored. A
endothelial cells (VEGFR), pericytes (PDGFR), and tumor randomized trial of a continuous dose protocol of 37.5 mg
cells (EGFR), and intracellular events contributing to cell daily sunitinib vs. standard 50 mg daily for 4 weeks fol-
differentiation, proliferation, and survival [39]. Activation lowed by a 2-week rest is underway.
of tyrosine kinases is a central event in the development of
RCC, a finding that is currently being exploited for thera- Sorafenib
peutic benefit.
Sorafenib is an oral small molecule inhibitor of multiple
kinases including raf, the VEGFR2 and VEGFR3 receptor
Targeted therapy in RCC tyrosine kinases, PDGFR-␤, FLT-3, and c-kit [51]. A Phase
II randomized discontinuation trial of sorafenib 400 mg BID
More than 10 agents have recently demonstrated activity in patients with solid tumors at five participating centers
in renal cell carcinoma, reflecting the success of identifying was completed with over 484 patients accrued (202 patients
targets in critical pathways. Most of these agents have been with RCC) [52]. All patients were treated in an open-label
studied in advanced RCC with clear cell features, as this is fashion with sorafenib for 3 months and responses were
the predominant subtype. Experiences in the non-clear cell measured. Patients who had a reduction in tumor volume of
populations are anecdotal; however, Phase II trials for tar- 25% or more by WHO criteria continued on sorafenib in an
gets specific to pathways such as c-met are currently under- open-label fashion until disease progression. Patients with-
way. The molecular targets important in clear cell RCC out progression and less than 25% reduction were random-
include VEGF receptors, the mammalian target of rapamy- ized to continue sorafenib or initiate placebo in a double-
cin, and the VEGF molecule itself (Table 1). Two novel blinded fashion. These patients were restaged every 6
agents, sorafenib and sunitinib, have recently been approved weeks. Upon progression, the treatment assignment was
by the FDA for use in advanced renal cell carcinoma, with unblinded and patients on the observation arm were permit-
more approvals anticipated. ted to resume sorafenib until subsequent progression.
During the run-in period of this trial, 73 of 202 patients
Sunitinib had tumor shrinkage of 25% or more. Sixty-five patients
with stable disease at 12 weeks were randomly assigned to
Sunitinib malate is an oral multitargeted tyrosine kinase take either sorafenib [32] or placebo [33]. At 24 weeks, 50%
inhibitor with antitumor and antiangiogenic activity directed of the patients on sorafenib had stable disease and 18% of
through inhibition of PDGFR, VEGFR 1 and 3, KIT, FLT3, placebo treated patients remained stable with a significantly
the receptor stem cell factor (SCF), and RET [41– 45]. FDA longer median PFS of 24 weeks vs. 6 weeks (placebo) (P ⫽
approval of this agent was based on Phase III data from 0.0087). A randomized Phase III trial of sorafenib vs. pla-
treatment of gastrointestinal stromal tumors and two Phase cebo in patients with advanced RCC who had failed one
II trials in metastatic RCC [46 – 48]. In the first RCC study, prior systemic treatment in the past 8 months was recently
63 patients received sunitinib orally at 50 mg daily for 4 completed, and results of the first 769 (384 sorafenib/385
weeks with a 2-week rest period. Entry criteria included placebo) of a total of 905 patients randomized were pre-
patients with measurable disease and disease progression on sented at ASCO 2005 [53]. Over 81% of patients in both
one prior cytokine-based therapy. Ninety-two percent of arms had received prior cytokine therapy and over 93% in
patients had prior nephrectomy. The results of this study both arms had prior nephrectomy. Fifty-seven percent of
424 N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432

patients in each arm had greater than two organ sites of Other mTOR inhibitors are currently in development or
metastatic disease. The median PFS was 24 weeks in the in trial, including everolimus (RAD001). In a recent Phase
sorafenib group compared with 12 weeks in the placebo II study, 7of 25 patients with mRCC with some clear cell
group. Discontinuation of drug occurred in 9% of patients features had partial responses to everolimus (RAD001)
on sorafenib compared with 6% on placebo due to adverse [65]. Multiple everolimus and temsirolimus combination
events. A Phase III trial of first-line sorafenib vs. IFN-␣ in trials are ongoing.
advanced RCC has recently been completed and is under-
going survival analysis. Anticipating these results, sor- Bevacizumab
afenib, like sunitinib, has replaced interferon as first-line
therapy for advanced RCC. Although not currently approved by the FDA for use in
RCC, bevacizumab was the first antiangiogenic agent
Other multitargeted tyrosine kinase receptor inhibitors shown to have activity in this disease. High-dose bevaci-
zumab (10 mg/kg), a neutralizing antibody to vascular en-
Other agents, including AG013736 (axitinib) [34], dothelial growth factor, improved PFS in a Phase II trial and
GW786034 (pazopanib) [50], ABT-869 [36], and AZD2171 heightened awareness of the importance of the HIF-1 ␣
[54] have demonstrated striking activity in Phase I or II pathway and angiogenesis in the regulation of RCC. In a
trials. The specific targets of some of these agents are randomized, placebo-controlled, double-blind trial, bevaci-
reviewed in Table 1. The use of multiple agents with mul- zumab was administered at 3 or 10 mg/kg every 2 weeks in
tiple and overlapping targets has helped identify targets or 116 patients with mRCC (40 to placebo, 37 to low-dose
combinations of targets that may be most important for antibody, and 39 to high-dose antibody) [66]. A significant
disease control. Investigation of these agents is ongoing at prolongation in time to progression of disease based on
multiple sites worldwide. WHO criteria was observed in the high-dose antibody group
compared with the placebo group (4.8 vs. 2.5 months).
Temsirolimus There was a small difference, of borderline significance,
between the time to progression of disease in the low-
Inhibition of the mammalian target of rapamycin dose antibody group (3.0 months) and that in the placebo
(mTOR/FRAP), a pathway with a serine/threonine kinase group. There were four partial responses (10% response
that regulates mRNA translation, is another treatment strat- rate) and a prolongation of time to tumor progression in
egy in metastatic RCC [55,56]. Both the rapamycin analog patients who received the higher dose of bevacizumab.
temsirolimus and rapamycin bind to FK506 binding protein, This trial by Yang et al. also validated the use of pro-
which inhibits mTOR [57]. Temsirolimus’ interaction with gression free survival as an index of activity for the
mTOR prevents phosphorylation of 4E-BP1 (PHAS-I), al- antiangiogenic drugs in this disease [66].
lowing this protein to bind to eIF4E and thus inhibiting Based on these findings, two randomized Phase III trials
mRNA translation. In addition to causing G1 arrest, there of interferon with and without bevacizumab were completed
are data that temsirolimus inhibits HIF by these mechanisms in Europe and the United States. These include the
[58 – 60]. AVOREN Phase III trial of IFN ␣-2a ⫹ placebo vs. IFN
A randomized Phase II trial of temsirolimus treating 111 ␣-2a ⫹ bevacizumab in metastatic RCC results conducted
patients with mRCC at 25, 75, or 250 mg i.v. weekly [61] in Europe and the CALGB trial of interferon- ␣ or interfer-
demonstrated a median survival of 15 months in patients on-␣ plus anti-VEGF antibody conducted in the United
with intermediate and unfavorable Motzer risk factors [62]. States [67]. The results of these trials are expected to be
This led to a Phase III trial of temsirolimus (25 mg i.v. reported at ASCO 2007.
weekly) vs. temsirolimus (15 mg i.v. weekly) and interferon
␣ (6 million units subcutaneously TIW) vs. interferon alpha
alone (up to 18 million units subcutaneously TIW) first-line Toxicities of targeted therapies
in 626 patients with poor-risk features [63]. The results of
this trial presented at ASCO 2006 demonstrated a signifi- Multi-targeted tyrosine kinase inhibitors
cant improvement in median overall survival (OS) in pa-
tients in the temsirolimus arm alone of 10.9 months (vs. 7.3 As a class, the multi-targeted receptor tyrosine kinases
months interferon alone) and less toxicity. This trial was the have characteristic side effects. These include fatigue, skin
first to demonstrate an improvement in OS for a targeted and hair, cardiovascular, endocrine, gastrointestinal and
agent in RCC. Since patients with predominantly poor risk metabolic effects. As more patients are treated with these
features were included in this trial, it is likely that this agent agents, more is known about the extent of each side effect.
will be further developed for this population. Indeed, data An ongoing study, E1Y03, sponsored by the Eastern Coop-
exists that demonstrates expression of mTOR pathway tar- erative Oncology Group (ECOG), is characterizing the pres-
gets in poor risk groups such as sarcomatoid subtype his- ence of pharmacogenetic cyp3A4 polymorphisms and phar-
tology [64]. macokinetics of sunitinib and sorafenib within populations
N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432 425

Table 2
Toxicities of targeted therapies
Toxicity [68,69] Sunitinib (n ⫽ 169) Sorafenib (n ⫽ 451) Bevacizumab (n ⫽ 39) Temsirolimus (n ⫽ 212)
10 mg/kg

Fatigue Likely Likely Less likely Likely


Hand-foot syndrome Less likely Likely ND ND
Other rash Less likely Likely Likely Likely
Hypertension Less likely Less likely Likely ND
Edema Less likely ND ND Less likely
Dyspnea Less likely Less likely ND Likely
LVEF decline Rare ND ND ND
Prolonged QT interval Rare ND ND ND
Anorexia Likely Less likely ND Likely
Diarrhea Likely Likely ND Likely
Stomatitis Likely Less likely ND Likely
Nausea Likely Less likely ND Likely
Fever Less likely Less likely Less likely Less likely
Bleeding Less likely Less likely Less likely Rare
Thrombosis Rare Rare Rare ND
Hypothyroidism Less likely ND ND ND
High AST/ALT Less likely Less likely Less likely Less likely
High amylase/lipase Less likely Less likely ND ND
High cholesterol ND ND ND Likely
High triglycerides ND ND ND Likely
Low phosphorus Less likely Less likely ND Likely
Neutropenia Less likely Less likely ND Likely
Thrombocytopenia Less likely Less likely ND Likely
Lymphopenia Less likely Less likely ND Likely
GI perforation Rare Rare Rare ND

Likely ⬎20%; less likely ⱕ20%; rare but serious ⬍3%.


ND ⫽ not described.

of patients treated with these agents to help determine if Sunitinib


subsets of populations are more prone to side effects. Many
side effects can be attributed to specific receptors targeted. The most commonly reported side effects of sunitinib are
The most commonly reported side effects, occurring in nausea, rash, fatigue, hypertension, diarrhea, and stomatitis
more than 10% of patients are depicted in Table 2. [68]. There is a higher incidence of myelosuppression re-
Hypertension is a common side effect for both the mul- ported with sunitinib than sorafenib. Two side effects are
titargeted tyrosine kinase inhibitors and VEGF inhibitors, specific to inhibition of stem cell factor and c-kit, and thus
but has not been correlated to response or to elevation in reported with sunitinib (as well as GW786034 and
VEGF levels [70]. A rare but serious complication is re- AG013736) [77]. These include a yellowish discoloration to
versible posterior leukoencephalopathy syndrome [71], the skin and/or lightening of hair. Additionally, altered
characterized by hypertension, headache, lethargy, altered thyroid function tests, including hypothyroidism, have been
mental status, and visual loss. The cardiac effects of this reported in one publication to be as high as 62% and 36%,
class have not been well characterized but may be similar to respectively [78]. This is most likely related to sunitinib
other agents such as imatinib and traztuzimab [72,73]. targeting of the RET proto-oncogene.
There are numerous skin manifestations of the tyrosine A toxicity sometimes seen with sunitinib is left ventric-
kinase inhibitors [74]. The most troubling of these is the ular dysfunction. The left ventricular ejection fraction
hand-foot syndrome. Hand-foot syndrome is a different (LVEF) was measured in a number of sunitinib trials con-
variety than that reported with chemotherapy agents such as ducted and the overall incidence of this abnormality was
the fluorodeoxyuridines [75]. It is characterized by an initial 10% with only 1% of these grade 3. As this potential side
discomfort in the palmar and plantar surfaces followed by effect has not been analyzed in sorafenib trials, it is un-
painful callus formation, often at pressure points and the known if this is a class effect [79]. A Phase IV cardiac study
appearance of sores underneath the calluses. A diffuse mac- is ongoing in the ASSURE Trial, which will prospectively
ular rash can also occur, which tends to improve with time. characterize the incidence of decline in LVEF as well as any
Another side effect that appears to be commonly associated relationship to congestive heart failure, hypertension, or
with both drugs is splinter hemorrhages in the nailbeds [76]. other toxicities to sorafenib, sunitinib or placebo. A rare
Additionally, either curling or lightening of the hair and side effect is seizure, reported in 1% of patients on sunitinib.
changes in skin tone can occur [77]. This can occur from drug interaction affecting anticonvul-
426 N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432

sant therapy serum levels, in the presence of previously events in the Phase II trial of everolimus were similar and
undetected brain metastases and idiopathically. Other rare included mucositis, skin rash, pneumonitis, hypophos-
side effects are thrombotic and bleeding complications, phatemia, hyperglycemia, thrombocytopenia, anemia, and
which are listed as rare but serious in Table 2. elevated LFTs [65]. Hypersensitivity reactions including
facial flushing and, rarely, difficulty breathing or speaking,
Sorafenib usually during the initial intravenous infusion have been
reported for temsirolimus [69].
The most frequently observed sorafenib-related toxicities Similar to organ-transplant patients receiving sirolimus,
include hand-foot skin reaction, rash, anorexia, diarrhea, pneumonitis/pulmonary infiltrates and alveolitis has been
fatigue, and hypertension [68]. Other common side effects reported among patients receiving intravenous temsirolimus
include pruritis, joint pain, and liver function abnormalities. [81]. Some patients have been asymptomatic with pneumo-
Neutropenia, thrombocytopenia, and anemia are uncom- nitis detected on CT scan or chest X-ray, while others have
mon, but not rare. The incidence of bleeding, thrombosis, had symptoms of dyspnea, cough, and fever. This side effect
and other serious side effects such as myocardial infarction appears to respond to steroid treatment and discontinuation
are reported as rare. A side effect specific to sorafenib is of the temsirolimus.
curling of the hair. Alopecia is also more common with Hyperlipidemia and hypophosphatemia are additional
sorafenib than sunitinib. Additionally, the incidences of class-specific toxicities of these agents secondary to mTOR
hypertension and hand-foot appear to be more common with inhibition [82,83]. Interference with insulin signaling path-
sorafenib than with sunitinib. ways, as a consequence of mTOR inhibition, appears to be
the mechanism responsible for these side effects [84]. The
Bevacizumab hyperlipidemia usually requires treatment with a lipid-low-
ering agent. Additionally, myelosuppression occurs com-
Most of the toxicity data reported for bevacizumab is monly when this agent is combined with cytotoxic agents.
reported from trials that incorporate bevacizumab with cy-
totoxic chemotherapy [68]. As such, many side effects may
have been reported in both chemotherapy alone and chemo- Targeted adjuvant strategies in RCC
therapy plus bevacizumab arms and can be attributed to
both the chemotherapy and antiangiogenic agents. Side ef- Between the extremes of early incidental RCC and
fects specific to bevacizumab alone can be singled out in a mRCC exists a gradation of risk. Twenty to 40% of patients
small Phase II trial. In the Phase II trial of high dose undergoing surgical resection for localized RCC experience
bevacizumab vs. low dose bevacizumab vs. placebo in ad- recurrence, suggesting that there are some individuals in
vanced RCC, toxic effects were minimal, with hypertension whom surgical excision is necessary but insufficient [4]. In
(35%) and proteinuria (64%) reported as the most substan- these patients, the development of effective adjuvant strat-
tial events in the high dose arm [66]. egies is imperative. The use of adjuvant therapies to treat
Potential side effects of bevacizumab plus chemotherapy patients with high-risk malignancies has been explored for
include gastrointestinal perforation, hemorrhage, impaired many different solid tumors. These efforts seek to eradicate
wound healing, and arterial thrombosis. The incidence of micrometastatic disease that has escaped surgical control
gastrointestinal perforation (2%) was best characterized in using systemic therapies. Due to poor outcomes for patients
the colorectal cancer population [68]. Hemorrhage has been with mRCC, studies have been undertaken to determine the
classified as minor, usually epistaxis, and major, including potential utility of various forms of adjuvant therapy for
hemoptysis in squamous cell lung cancer (31%), hemopty- those patients at high risk for recurrent disease. Historically,
sis in adenocarcinoma of the lung (4%), CNS bleeding postoperative adjuvant therapies have not been shown to
(hemorrhagic stroke and subarachnoid hemorrhage, uncom- have significant clinical benefit for these patients [85]. In
mon) [80]. Arterial thrombotic events (cerebral infarction, spite of this, the initial time after surgical resection holds
transient ischemic attacks, myocardial infarction, angina, promise as a period with minimal tumor bulk corresponding
and others), pooled from 1,745 patients in randomized trials with a newly “unsuppressed” immune system whereby ad-
of chemotherapy with and without the bevacizumab label, juvant strategies may be successful.
had an incidence of 1.9% vs. 4.4%, respectively [68].
Published adjuvant trials in RCC
mTOR inhibitors
Agents that have been studied as adjuvant treatment for
The rapamycin analogs have some class-specific side high risk RCC include radiotherapy, hormonal therapies,
effects as well. The most common side effects of the tem- and immunotherapies. Unfortunately, nearly all the trials
sirolimus in a Phase 2 study of 110 patients with RCC are examining the use of local or systemic therapies have failed
rash (72%), mucositis (65%), asthenia (39%), nausea to demonstrate any benefit in the adjuvant setting [85].
(36%), and acne (30%) [61]. Treatment related adverse Among these trials, the most significant failures have all
N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432 427

been using immunotherapeutic strategies, which for nearly nephrectomy for 612 patients with high-risk nonmetastatic
two decades has been the standard of care for patients with clear cell RCC. This trial is currently accruing patients.
mRCC. Multiple trials examining the effects of adjuvant On the basis of the aforementioned favorable data in
interferon-␣, recombinant interferon ␣-2b, autologous irra- patients with mRCC using the oral tyrosine kinase inhibitors
diated tumor cells mixed with BCG, IL-2, subcutaneous sorafenib and sunitinib, a Phase III trial is currently under-
cytokines with 5-FU, and combination IL-2/FN-2␣/5-FU way randomizing patients to receive either sorafenib,
have all failed to show any survival advantages over obser- sunitinib, or placebo after definitive resection of localized or
vation alone [85,86]. locally advanced RCC. The study, termed ASSURE (adju-
To our knowledge there has only been a single positive vant sorafenib sunitinib for unfavorable renal cell carci-
prospective adjuvant randomized trial in RCC. In a multi- noma), is sponsored by ECOG and hopes to accrue 1,332
center Phase III trial using individually prepared autologous patients over 4 years. Eligible patients must have fully
renal tumor cell vaccine injected intradermally at 55 Ger- resected, locally advanced, nonmetastatic disease with or
man centers following radical nephrectomy, 5-year PFS was without lymph node involvement. Progression-free survival
77.4% following vaccine vs. 67.8% in the observation arm is the primary endpoint. Patients are registered for this trial
(P ⫽ 0.02, log-rank test) [87]. Importantly, 32% of enrolled based on unfavorable pathologic findings after surgical re-
patients (174/553) were lost after randomization with a section or preoperatively based on high-risk radiographic
disproportionate number of losses from the treatment group. findings, thus allowing for tissue correlative studies. Sec-
Moreover, OS differences were not analyzed, raising sig- ondary endpoints include overall survival and toxicity in the
nificant questions regarding the interpretation and applica- adjuvant setting.
bility of these results. Another Phase III trial examining the prolonged use of
sorafenib in the adjuvant setting sponsored by the Medical
Adjuvant targeted trials in RCC Research Council (MRC) is currently planned. SOrafenib in
Renal CEll (SORCE) is a randomized controlled study com-
Several important trials currently underway are examin- paring sorafenib administered for 3 years or 1 year with
ing adjuvant, primarily targeted agents in high risk but fully placebo in patients with resected primary renal cell carci-
surgically resected RCC. The European Organization for noma at high or intermediate risk of relapse. This study aims
Research and Treatment of Cancer (EORTC) has coordi- to recruit approximately 331 patients per year for each of 5
nated a Phase III adjuvant trial to further evaluate interleu- years with an additional 3 years of follow-up. The primary
kin-2, interferon-␣, and 5-fluorouracil for patients with high endpoint is metastasis-free survival and secondary measures
risk of relapse after surgical treatment for RCC (EORTC include CSS, overall survival, cost effectiveness, and tox-
protocol 30955). In this study, 550 patients have been ran- icity [85,86] (Table 3).
domized to either treatment or observation after surgery.
Now closed to recruitment, analysis will determine disease-
free survival and overall survival in each arm [86]. Surgical considerations of targeted therapies in RCC
RCC is highly vascular with disordered angiogenesis.
Due to its antiangiogenic effects, thalidomide has been As targeted therapies in RCC have been introduced into
studied in mRCC in a Phase III trial for patients with stage urologic clinics, several practical and biologically relevant
IV disease randomized patients to receive interferon-␣ with surgical issues have been raised as the collective experience
or without thalidomide. This study demonstrated an im- with these agents grows. For example, since these agents
provement in progression-free survival using thalidomide target tissue vascularity, do they have any definable adverse
[88]. Based on these findings, an adjuvant study was initi- effects on intra- or postoperative bleeding? What are the
ated at M.D. Anderson for patients with high risk RCC implications of these agents on perioperative wound heal-
comparing thalidomide to controls after nephrectomy. This ing? How should patients taking these agents be advised of
Phase II trial had an accrual goal of 220 patients but has management prior to elective surgery? Are there other clin-
closed due to poor accrual. ical urologic concerns? Whereas these agents have only
A currently ongoing clinical trial involves the use of a been approved for use outside of clinical trials since De-
monoclonal antibody against G250, a transmembrane pro- cember 2005, data of this nature remain limited.
tein associated with RCC, which is identical to CAIX [74].
cG250 (WX-G250, Rencarex®; Wilex Pharmaceuticals, Neoadjuvant targeted therapies and the role of
Munich, Germany) is a monoclonal IgG1 antibody admin- cytoreduction
istered intravenously that binds to CAIX on clear cell RCC
and may recruit effector cells or activate complement to Since nearly 4 in 10 patients with RCC present with
result in cell death [86]. In 2004, an international random- locally advanced or metastatic disease [1], the role of sur-
ized, Phase III trial termed ARISER (adjuvant rencarex gical removal of the primary tumor in advanced disease has
immunotherapy trial to study efficacy in nonmetastatic been debated and reviewed [89]. Arguments for cytoreduc-
RCC) opened to accrual to evaluate cG250 vs. placebo after tion include a small body of literature demonstrating spon-
428
Table 3
Current adjuvant trials for high-risk RCC
Trial Sponsor Study groups Route Study design Treatment Accrual Inclusion criteria Status
duration goal

N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432
ARISER, Rencarex (G250 mAb) Wilex G250 mAb vs. i.v. Phase III, double- 24 weeks 600 ● Histologically confirmed primary Open and accruing
placebo blind, placebo- clear cell RCC
controlled ● Meets one of the following stage
criteria:
- stage T3a, T3b, T3c, or T4 and
No/NxMo
- any T stage and N⫹ M0
- T1bN0/NXM0 or T2N0/NXM0
with microscopic vascular inva-
sion and grade ⱖIII or T3aN0/
NX°M0 with grade ⱖIII
● Fully resected within 12 weeks
● No evidence of macroscopic or
microscopic residual disease
ASSURE, sorafenib/sunitinib ECOG Sunitinib vs. sorafenib PO Phase III, randomized, 1 year 1,332 ● Patients with high-grade (G3) Open and accruing
vs. placebo for 1 double-blind, T1b and any grade with a higher
year placebo-controlled T stage (T2-4)
● Patients with Nx, N0, N1, or N2
if all positive nodes were
removed
● Patients must be M0
● Patients can have a laparoscopic
or open, partial or radical
● No more than 3months since
surgery
● No evidence of residual or
recurrent disease
● Clear cell and non-clear cell
tumors eligible
SORCE, sorafenib MRC Sorafenib (for 1 or 3 PO Phase III, three-arm, 3 years 1,650 ● Intermediate- or high-risk disease Activated May 2007
years) vs. placebo double-blind, RCC (clear cell and non-clear
controlled cell)
● No more than 3 months prior to
treatment start date
● No evidence of residual
macroscopic disease after
resection
N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432 429

taneous regression of metastatic sites upon removal of the zumab 20 days, sunitinib and sorafenib 5 days, temsirolimus
primary tumor, reduction of the overall tumor burden in an 15 hours) are helpful in planning surgical procedures, and it
effort to make systemic therapies more effective, alleviation is generally recommended to discontinue bevacizumab 4
of symptoms or the prevention of future symptoms due to weeks prior to surgery. The reported incidence of wound
the primary tumor, and removal of tissues for clinical trials. healing complications in patients who had taken bevaci-
Arguments against radical nephrectomy in the setting of zumab 28 to 60 days prior to surgery was 1.3% vs. 0.5% in
metastatic disease include the definable morbidity and mor- non-bevacizumab treated patients and 13% vs. 3.4%, re-
tality of surgery in elderly, higher risk patients and the spectively, for complications from surgery while on therapy
associated postsurgical delay in beginning systemic thera- in one series [92]. The same series reported surgical com-
pies. plications in patients receiving bevacizumab within less
Nonetheless, current practice is to perform cytoreduc- than 30 and 30 to 60 days after surgery. The authors con-
tive nephrectomy when possible. This is based primarily cluded that receiving bevacizumab within 28 days of sur-
on the results of two randomized clinical trials demon- gery was safe and feasible. Another study recommended a
strating a statistically significant 6 month survival benefit window of 6 to 8 weeks postoperatively following bevaci-
to cytoreduction and IFN vs. IFN alone [14,90]. It is zumab therapy [93].
therefore important to note that there has not been a Information in the product labels and PDR for sorafenib
demonstrated benefit to cytoreduction in the targeted and sunitinib is limited and recommends physician discre-
therapy era. Recently a RAND appropriateness survey tion. Clearly, there are anecdotes of delayed wound healing
was performed to evaluate data regarding treatment of with minor injuries such as abrasions using these agents,
mRCC including the role of cytoreduction [91]. A total of and the common entry criterion for both sunitinib and sor-
22 controlled trials were reviewed (20 prospective) in- afenib based trials includes a minimum of 4 weeks post
cluding 4,289 patients with mRCC. Cytoreduction was major surgery prior to initiating therapy; however, given the
deemed most appropriate for patients with good surgical shorter half-lives of these agents, the use of these agents
risk, symptoms related to the primary tumor, and limited preceding or following surgery may be more permissive.
metastatic burden. For patients with planned immuno or Little data exist for the impact of temsirolimus on wound
targeted therapy and good surgical risk status, cytoreduc- healing or surgical complications. Given that this target is
tion was also deemed appropriate in patients with limited upstream to the immediate VEGF pathways, it is possible
metastatic burden and no primary tumor-related symp- that the wound and surgical issues are of less importance
toms. In good surgical risk patients with extensive tumor with this agent. More data are needed with temsirolimus as
burden with symptoms, cytoreduction was deemed ap- well as the other agents in this class.
propriate in patients who were planning postoperative
immunotherapy, but its role was uncertain in those same
patients planning postoperative targeted therapy. The role
Future directions
of cytoreductive nephrectomy was considered uncertain
or inappropriate for poor surgical risk patients and those
The use of targeted therapies for RCC is a direct result of
without primary tumor-related symptoms who had exten-
refinements in basic science techniques and our understand-
sive metastatic burdens [91]. These findings underscore
ing of the complexities of the VEGF pathway. Ongoing
the uncertainty associated with primary tumor responses
Phases I and II trials will address the use of these agents in
following targeted therapy, particularly for asymptom-
combination as parallel or sequential targeted agents. From
atic, poor risk patients with extensive metastatic burdens.
these studies, more data will be compiled about the rele-
Although several institutionally based clinical trials are
vance of the particular targets to toxicities as well as to
currently underway examining the clinical and biological
response. Ongoing studies hope to refine the use of these
responses of the primary tumor and their associated me-
agents in appropriate populations. Many questions remain.
tastases to targeted therapies, until these data become
Nonetheless, as the pathways responsible for RCC are better
available, the clinician should select patients for cytore-
understood, the role of targeted therapies in RCC will con-
duction carefully in the targeted therapy era.
tinue to expand.
Wound healing and other concerns following targeted
therapies
References
The risks of bleeding, hemorrhage, and gastrointestinal
perforation have best been characterized in bevacizumab, [1] Jemal A, Murray T, Ward E, et al. Cancer statistics, 2005. CA Cancer
J Clin 2005;55:10 –30.
but data are emerging for the multitargeted tyrosine kinase
[2] Cohen HT, McGovern FJ. Renal-cell carcinoma. N Engl J Med
inhibitors. Several publications have emerged addressing 2005;353:2477–90.
the safety of the use of bevacizumab in association with [3] Jayson M, Sanders H. Increased incidence of serendipitously discov-
surgical procedures [92,93]. The serum half-lives (bevaci- ered renal cell carcinoma. Urology 1998;51:203–5.
430 N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432

[4] Lam JS, Shvarts O, Leppert JT, et al. Renal cell carcinoma 2005: New [28] Hatano N, Nishikawa NS, McElgunn C, et al. A comprehensive
frontiers in staging, prognostication, and targeted molecular therapy. analysis of loss of heterozygosity caused by hemizygous deletions in
J Urol 2005;173:1853– 62. renal cell carcinoma using a subtraction library. Mol Carcinog 2001;
[5] Fuhrman SA, Lasky LC, Limas C. Prognostic significance of mor- 31:161–70.
phologic parameters in renal cell carcinoma. Am J Surg Path 1982; [29] Schmidt L, Duh FM, Chen F, et al. Germline and somatic mutations
6:655– 63. in the tyrosine kinase domain of the MET proto-oncogene in papillary
[6] Lam JS, Leppert JT, Figlin RA, et al. Role of molecular markers in renal carcinomas. Nat Genet 1997;16:68 –73.
the diagnosis and therapy of renal cell carcinoma. Urology 2005; [30] Speicher MR, Schoell B, du Manoir S, et al. Specific loss of chro-
66(Suppl 5A):1–9. mosomes 1, 2, 6, 10, 13, 17, and 21 in chromophobe renal cell
[7] Gonzalgo MR, Yegnasubramanian S, Yan G, et al. Molecular profil- carcinomas revealed by comparative genomic hybridization. Am J
ing and classification of sporadic renal cell carcinoma by quantitative Pathol 1994;145:356 – 64.
methylation analysis. Clin Cancer Res 2004;10:7276 – 83. [31] van den Berg E, van der Hout AH, Oosterhuis JW, et al. Cytogenetic
[8] Chawla SN, Crispen PL, Hanlon AL, et al. The natural history of analysis of epithelial renal-cell tumors: Relationship with a new
observed enhancing renal masses: Meta-analysis and review of the histopathologic classification. Int J Cancer 1993;55:223–7.
world literature. J Urol 2006;175:425–31. [32] Kenck C, Wilhelm M, Bugert P, et al. Mutation of the VHL gene is
[9] Frank I, Blute ML, Leibovich BC, et al. Independent validation of the associated exclusively with the development of nonpapillary renal
2002 American Joint Committee on cancer primary tumor classifica- cell carcinomas. J Pathol 1996;179:157– 61.
tion for renal cell carcinoma using a large, single institution cohort. [33] Fuzesi L, Cober M, Mittermayer C. Collecting duct carcinoma: Cy-
J Urol 2005;173:1889 –92. togenetic characterization. Histopathology 1992;21:155– 60.
[10] Moinzadeh A, Gill IS, Finelli A, et al. Laparoscopic partial nephrec- [34] Rini B, Rixe O, Bukowski R, et al. AG-013736, a multi-target
tomy: Three-year follow-up. J Urol 2006;175:459 – 62. tyrosine kinase receptor inhibitor, demonstrates antitumor activity in
[11] Gill IS, Remer EM, Hasan WA, et al. Renal cryoablation: Outcome at a Phase 2 study of cytokine-refractory, metastatic renal cell cancer
three years. J Urol 2005;173:1903–7. (RCC). Proceedings of the ASCO 2005 Annual Meeting. J Clin Oncol
[12] Yagoda A, Petrylak D, Thompson S. Cytotoxic chemotherapy for 2005;23(Suppl) [Abstract 4509].
advanced renal cell carcinoma. Urol Clin North Am 1993;20:303–21. [35] Hurwitz H, Dowlati A, Savage S, et al. Safety, tolerability, and
[13] Motzer RJ, Russo P. Systemic therapy for renal cell carcinoma: J Urol pharmacokinetics of oral administration of GW786034 in patients
2000;163:408 –17. with solid tumors. Proceedings of the ASCO 2005 Annual Meeting.
[14] Flanigan RC, Salmon SE, Blumenstein BA, et al. Nephrectomy fol- J Clin Oncol 2005;23(Suppl) [Abstract 3012].
lowed by interferon ␣-2b compared with interferon ␣-2b alone for [36] Albert DH, Tapang P, Magoc TJ, et al. Preclinical activity of ABT-
metastatic renal cell cancer. N Engl J Med 2001;345:1655–9. 869, a multitargeted receptor tyrosine kinase inhibitor. Mol Cancer
[15] Motzer RJ, Bander NH, Nanus DM. Renal-cell carcinoma. N Engl Ther 2006;5:995–1006.
J Med 1996;335:865–75. [37] Wood JM, Bold G, Buchdunger E, et al. PTK787/ZK 222584, a novel
[16] Coppin C, Porzsolt F, Awa A, et al. Immunotherapy for advanced and potent inhibitor of vascular endothelial growth factor receptor
renal cell carcinoma. Cochrane Database Syst Rev 2005;25: tyrosine kinases, impairs vascular endothelial growth factor-induced
CD001425. responses and tumor growth after oral administration. Cancer Res
[17] Enquist E, Zambrano N, Zbar B, et al. Molecular genetics of renal cell 2000;60:2178 – 89.
carcinoma. In: Bukowski RM, Novick AC, editors. Renal cell carci- [38] Drevs J, Muller-Driver R, Wittig C, et al. PTK787/ZK 222584, a
noma: Molecular biology, immunology, and clinical management. specific vascular endothelial growth factor-receptor tyrosine kinase
Totowa (NJ): Humana Press Inc., 2000. p. 79 –93. inhibitor, affects the anatomy of the tumor vascular bed and the
[18] Knudson AG Jr. Genetics of human cancer. Annu Rev Genet 1986; functional vascular properties as detected by dynamic enhanced mag-
20:231–51. netic resonance imaging. Cancer Res 2002;62:4015–22.
[19] Uzzo RG, Novick AC. von Hippel-Lindau syndrome: Clinical and [39] Stadler WM. Targeted agents for the treatment of advanced renal cell
molecular considerations for the urologist. AUA Update Series 1999; carcinoma. Cancer 2005;104:2323–33.
18:137– 44. [40] Cohen HT, McGovern FJ. Renal cell carcinoma. N Engl J Med
[20] Lubensky IA, Schmidt L, Zhuang Z, et al. Hereditary and sporadic 2005;353:2477–90.
papillary renal carcinomas with c-met mutations share a distinct [41] Mendel DB, Laird AD, Xin X, et al. In vivo antitumor activity of
morphological phenotype. Am J Pathol 1999;155:517–26. SU11248, a novel tyrosine kinase inhibitor targeting vascular endo-
[21] Uzzo RG, Cairns P, Al-Saleem T, et al. The basic biology and thelial growth factor and platelet-derived growth factor receptors:
immunobiology of renal cell carcinoma: Considerations for the cli- Determination of a pharmacokinetic/pharmacodynamic relationship.
nician. Urol Clin N Am 2003;30:423–36. Clin Cancer Res 2003;9:327–37.
[22] Baylin SB, Herman JG, Graff JR, et al. Alterations in DNA methyl- [42] O’Farrell AM, Abrams TJ, Yuen HA, et al. SU 11248 is a novel FLT3
ation: A fundamental aspects of neoplasia. Adv Cancer Res 1998;72: tyrosine kinase inhibitor with potent activity in vitro and in vivo.
141–96. Blood 2003;101:3597– 605.
[23] Seizinger BR, Rouleau GA, Ozelius LJ, et al. von Hippel-Lindau [43] Rosen L, Mulay M, Long J, et al. Cancer Institute Medical Group,
disease maps to the region of chromosome 3 associated with renal cell Santa Monica, CA; Sugen/Pharmacia, San Francisco, CA. Phase I
carcinoma. Nature 1988;332:268 –9. trial of SU011248, a novel tyrosine kinase inhibitor in advanced solid
[24] Gnarra JR, Lerman MI, Zbar B, et al. Genetics of renal cell carcinoma tumors. Proc Am Soc Clin Oncol 2003;22:191 [Abstract 765].
and evidence for a critical role for von Hippel-Lindau in renal tu- [44] Raymond E, Faivre S, Vera K, et al. For the SU11248 working group;
morigenesis. Semin Oncol 1995;22:3– 8. Institute Gustave Roussy, Villejuif, France; Pharmacia Co. and Sugen
[25] Maher ER, Kaelin WG. von Hippel-Lindau disease. Medicine 1997; Inc., Villejuif, France. Final results of a Phase I and pharmacokinetic
76:381–91. study of SU11248, a novel multi-target tyrosine kinase inhibitor, in
[26] Linehan WM, Lerman MI, Zbar B. Identification of the von Hippel- patients with advanced cancers. Proc Am Soc Clin Oncol 2003;22:
Lindau (VHL) gene. Its role in renal cancer. JAMA 1995;273: 192 [Abstract 769].
564 –70. [45] Fiedler W, Serve H, Döhner H, et al. A Phase I study of SU11248 in
[27] Foster K, Prowse A, van den Berg A, et al. Somatic mutations of the the treatment of patients with refractory or resistant acute myeloid
von Hippel-Lindau disease tumor suppressor gene in nonfamilial leukemia or not amenable to conventional therapy for the disease.
clear cell renal carcinoma. Hum Mol Genet 1994;3:2169 –73. Blood 2005;105:986–93.
N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432 431

[46] Motzer RJ, Rini BI, Bukowski RM, et al. Sunitinib in patients with the ASCO 2006 Annual Meeting. Part I. J Clin Oncol 2006;24(Suppl)
metastatic renal cell carcinoma. JAMA 2006;295:2516 –24. [Abstract LBA4].
[47] Motzer RJ, Rini BI, Michaelson MD, et al. SU011248, a novel [64] Figlin RA, Seligson D, Wu H, et al. Characterization of the mTor
tyrosine kinase inhibitor, shows antitumor activity in second line pathway in renal cell carcinoma and its use in predicting patient
therapy for patients with metastatic renal cell carcinoma: Results of a selection for agents targeting this pathway. Proceedings of the ASCO
Phase II study. Proc Am Soc Clin Oncol 2004;23:381 [Abstract 2005 Annual Meeting, Part I. J Clin Oncol 2005;23(Suppl) [Abstract
4500]. 4539].
[48] Demetri GD, van Oosterom AT, Garrett CR, et al. Efficacy and safety [65] Amato RJ, Misellati A, Khan M, et al. A Phase II trial of RAD001 in
of sunitinib in patients with advanced gastrointestinal stromal tumor patients with metastatic renal cell carcinoma (MRCC). J Clin Oncol
after failure of imatinib: A randomized controlled trial. Lancet 1006; 2006 Proceedings of the ASCO 2006 Annual Meeting, Part I. 2006;
368:1329 –38. 24(Suppl) [Abstract 4530].
[49] Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to [66] Yang JC, Haworth L, Sherry RM, et al. A randomized trial of
evaluate the response to treatment in solid tumors. European Orga- bevacizumab, an antivascular endothelial growth factor antibody for
nization for Research and Treatment of Cancer, National Cancer metastatic renal cancer. N Engl J Med 2003;349:427–34.
Institute of the United States, National Cancer Institute of Canada. [67] Rini B, Halabi S, Taylor J, et al. Cancer and Leukemia Group B
J Natl Cancer Inst 2002;92:205–16. 90206: A randomized Phase III trial of interferon-␣ or interferon-␣
[50] Motzer RJ, Hutson TE, Tomczak P, et al. Phase III randomized trial plus antivascular endothelial growth factor antibody (bevacizumab)
of sunitinib malate (SU11248) vs. interferon-␣ (IFN-␣) as first-line in metastatic renal cell carcinoma. Clin Cancer Res 2004;10:2584 – 6.
systemic therapy for patients with metastatic renal cell carcinoma [68] Physicians’ Desk Reference (PDR), 60th edition. Montvale (NJ):
(mRCC). Proceedings of the ASCO 2006 Annual Meeting, Part I. Medical Economics Co., 2006.
J Clin Oncol 2006;24(Suppl) [Abstract LBA3]. [69] Investigator Safety Report for CCI-779. Investigator’s Brochure.
[51] Wilhelm SM, Carter C, Tang L, et al. BAY 43-9006 exhibits broad April, 2005.
spectrum oral antitumor activity and targets the RAF/MEK/ERK [70] Veronese ML, Mosenkis A, Flaherty KT, et al. Mechanisms of
pathway and receptor tyrosine kinases involved in tumor progression hypertension associated with BAY 43-9006. J Clin Oncol 2006;24:
and angiogenesis. Cancer Res 2004;64:7099 –109. 1363–9.
[52] Ratain MJ, Eisen T, Stadler WM, et al. Phase II placebo-controlled [71] Govindarajan R, Adusumilli J, Baxter DL, et al. Reversible posterior
randomized discontinuation trial of sorafenib in patients with meta- leukoencephalopathy syndrome induced by RAF kinase inhibitor
static renal cell carcinoma. J Clin Oncol 2006;24:2505–12. BAY 43–9006. J Clin Oncol 2006;24:e48.
[53] Escudier B, Szczylik C, Eisen T, et al. Randomized Phase III trial of [72] Ewer MS, Vooletich MT, Durand JB, et al. Reversibility of trastu-
the RAF kinase and VEGFR inhibitor sorafenib (BAY 43-9006) in zumab-related cardiotoxicity: New insights based on clinical course
patients with advanced renal cell carcinoma (RCC). Proc Am Soc and response to medical treatment. J Clin Oncol 2005;23:7820 – 6.
Clin Oncol 2005;23 [Abstract 4510]. [73] Ewer MS, Lippman SM. Type II chemotherapy-related cardiac dys-
[54] Van Cruijsen H, Voest EE, Van Herpen CM, et al. Phase I evaluation function: Time to recognize a new entity. J Clin Oncol 2005;23:
of AZD2171, a highly potent, selective VEGFR signaling inhibitor, in 2900 –2.
combination with gefitinib, in patients with advanced tumors. Pro- [74] Robert C, Soria JC, Spatz A, et al. Cutaneous side-effects of kinase
ceedings of the ASCO 2006 Annual Meeting. J Clin Oncol 2006; inhibitors and blocking antibodies Lancet Oncol 2005;6:491–500.
24(Suppl) [Abstract 3017]. [75] Lokich JJ, Moore C. Chemotherapy-associated palmar-plantar eryth-
[55] Albers MW, Williams RT, Brown EJ, et al. FKBP-rapamycin inhibits rodysesthesia syndrome. Ann Intern Med 1984;101:798 –9.
a cyclin-dependent kinase activity and a cyclin D1-Cdk association in [76] Robert C, Faivre S, Raymond E, et al. Subungual splinter hemor-
early G1 of an osteosarcoma cell line. J Biol Chem 1993;268: rhages: A clinical window to inhibition of vascular endothelial
22825–9. growth factor receptors? Ann Intern Med 2005;143:313– 4.
[56] Hidalgo M, Rowinsky EK. The rapamycin-sensitive signal transduc- [77] Routhouska S, Gilliam AC, Mirmirani P. Hair depigmentation during
tion pathway as a target for cancer therapy. Oncogene 2000;19: chemotherapy with a class III/V receptor tyrosine kinase inhibitor.
6680 – 6. Arch Dermatol 2006;142:1477–9.
[57] Brown EJ, Albers MW, Shin TB, et al. A mammalian protein targeted [78] Desai J, Yassa L, Marqusee E, et al. Hypothyroidism after sunitinib
by G1-arresting rapamycin receptor complex. Nature (Lond) 1994; treatment for patients with gastrointestinal stromal tumors. Ann Intern
369:756 – 8. Med 2006;145:660 – 4.
[58] Dudkin L, Dilling MB, Cheshire PJ, et al. Biochemical correlates of [79] Ewer MS, Lippman SM. Type II chemotherapy-related cardiac dys-
mTOR inhibition by the rapamycin ester CCI-779 and tumor growth function: Time to recognize a new entity. J Clin Oncol 2005;23:
inhibition. Clin Cancer Res 2001;7:1758 – 64. 2900 –2.
[59] Neshat MS, Mellinghoff IK, Tran C, et al. Enhanced sensitivity of [80] Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or
PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad with bevacizumab for non-small-cell lung cancer. N Engl J Med
Sci USA 2002;98:10314 –9. 2006;355:2542–50.
[60] Del Bufalo D, Ciuffreda L, Trisciuoglio D, et al. Antiangiogenic [81] Duran I, Sin LL, Oza Am, et al. Characterization of the lung
potential of the mammalian target of rapamycin inhibitor temsiroli- toxicity of the cell cycle inhibitor temsirolimus. Eur J Cancer
mus. Cancer Res 2006;66:5549 –54. 2006;42:1875– 80.
[61] Atkins MB, Hidalgo M, Stadler WM, et al. Randomized Phase II [82] Galanis E, Buckner JC, Maurer MJ, et al. Phase II trial of temsiroli-
study of multiple dose levels of CCI-779, a novel mammalian target mus (CCI-779) in recurrent glioblastoma multiforme: A North Cen-
of rapamycin kinase inhibitor, in patients with advanced refractory tral Cancer Treatment Group Study. J Clin Oncol 2005;23:5294 –304.
renal cell carcinoma. J Clin Oncol 2004;22:909 –18. [83] Chang SM, Wen P, Cloughesy T, et al. Phase II study of CCI-779 in
[62] Motzer RJ, Bacik J, Schwartz LH, et al. Prognostic factors for patients with recurrent glioblastoma multiforme. Invest New Drugs
survival in previously treated patients with metastatic renal cell car- 2005;23:357– 61.
cinoma. J Clin Oncol 2004;22:454 – 63. [84] Huffman TA, Mothe-Satney I, Lawrence JC Jr. Insulin-stimulated
[63] Hudes G, Carducci M, Tomczak P, et al. A Phase 3, randomized, phosphorylation of lipin mediated by the mammalian target of rapa-
three-arm study of temsirolimus (TEMSR) or interferon-␣ (IFN) or mycin. Proc Natl Acad Sci USA 2002;99:1047–52.
the combination of TEMSR ⫹ IFN in the treatment of first-line, [85] Kunkle DA, Haas NB, Uzzo RG. Adjuvant therapy for high-risk renal
poor-risk patients with advanced renal cell carcinoma. Proceedings of cell carcinoma patients. Curr Urol Rep 2007;8:19 –30.
432 N.B. Haas, R.G. Uzzo / Urologic Oncology: Seminars and Original Investigations 25 (2007) 420 – 432

[86] Lam JS, Leppert JT, Belldegrun AS, et al. Adjuvant therapy of renal [90] Mickisch Garin A, van Poppel H, de Prijck L, et al. European
cell carcinoma: Patient selection and therapeutic options. BJU Int Organization for Research and Treatment of Cancer (EORTC)
2005;96:483– 8. Genitourinary Group. Radical nephrectomy plus interferon-␣-
[87] Jocham D, Richter A, Hoffman L, et al. Adjuvant autologous renal based immunotherapy compared with interferon ␣ alone in meta-
tumor cell vaccine and risk of tumour progression in patients with static renal-cell carcinoma: A randomized trial. Lancet 2001;358:
renal-cell carcinoma after radical nephrectomy: Phase III randomized 966 –70.
controlled trial. Lancet 2004;363:594 –9. [91] Halbert RJ, Figlin RA, Atkin MB, et al. Treatments for patients with
[88] Gordon MS, Manola J, Fairclough D, et al. Low dose interferon-␣2b metastatic renal cell cancer: A Rand Appropriateness Panel. Cancer
⫹ thalidomide in patients with previously untreated renal cell cancer: 1006;107:2375– 83.
Improvement in progression-free survival but not quality of life or [92] Scappaticci FA, Fehrenbacher L, Cartwright T, et al. Surgical wound
overall survival, a Phase III study of the Eastern Cooperative Oncology healing complications in metastatic colorectal cancer patients treated
Group (E2898). Proc Am Soc Clin Oncol 2004;22 [Abstract 4516]. with bevacizumab. J Surg Oncol 1005;91:173– 80.
[89] Uzzo RG, Novick AC. The role of nephrectomy and metastasectomy [93] D’Angelica M, Kornprat P, Gonen M, et al. Lack of evidence for
in advanced renal cell carcinoma. In: Bukowski RM, Novick AC, increased operative morbidity after hepatectomy with periopera-
editors. Renal cell carcinoma: Molecular biology, immunology, and tive use of bevacizumab: A matched case-control study. Ann Surg
clinical management. Totowa (NJ): Humana Press, 2000. p. 215–228. Oncol 2007;14:759 – 65.

Anda mungkin juga menyukai