Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Targ Oncol (2009) 4:121–133

DOI 10.1007/s11523-009-0109-x

REVIEW

Renal toxicity of targeted therapies


Ronan J. Kelly & Bertrand Billemont & Olivier Rixe

Received: 10 February 2009 / Accepted: 31 March 2009 / Published online: 6 May 2009
# US Government 2009

Abstract The use of molecular targeted therapies for the types of solid tumors have emerged. An improved
treatment of cancer has increased over the last decade. The understanding of the molecular biology involved in the
benefits of these compounds in terms of efficacy are often development of cancer has stimulated a dramatic increase in
relatively modest and counter balanced by the occurrence both research and development for novel antitumor thera-
of significant toxicities. Many of these newer agents used in pies. Several molecular targeted compounds have now been
clinical practice lack specificity and selectivity and have a approved by the US Food and Drug Administration and the
propensity to inhibit multiple targets. The biological European Medicines Agency for use as single modality or
consequences of multi-kinase activity are poorly defined in combination with conventional cytotoxic chemothera-
and numerous class-specific toxicities have been described. pies. Inhibition of the vascular endothelial growth factor
The kidney is an organ where most of these targeted (VEGF) and epidermal growth factor receptor (EGFR)
pathways are expressed. Preclinical data and human renal pathways have been the most successful directions to date
biopsies have generated an understanding of the mecha- in translating scientific knowledge to clinical benefit for the
nisms involved in how targeted agents can cause renal treatment of solid tumors [1].
toxicity. This review article discusses the observed nephro- Analysis of large phase II and III clinical trial data has
toxicity with this burgeoning class of therapeutics and exposed several limitations for the use of targeted agents.
reviews both the biological reasons for its occurrence and The demonstration of intrinsic and acquired resistance as
possible ways to prevent significant renal damage. well as the demonstration of specific toxicities involving
the skin, the cardiovascular system and the kidney have
Keywords Targeted therapies . Renal toxicity . Sunitinib . been reported [2]. These observations, in conjunction with
Bevacizumab . Sorafenib preclinical data, reveal the absence of specificity of these
newer agents. It has been suggested that co-expression of
membrane targets or shared intracellular pathways between
Introduction normal tissue and cancer cells may explain the occurrence
of the described adverse events (Fig. 1). Additional
Over the last 10 years, new treatment paradigms incorpo- mechanisms of toxicities are also suspected. Monoclonal
rating the use of targeted therapy in the treatment of many antibodies against VEGF or small molecule VEGF-
receptor (VEGFR) tyrosine kinase inhibitors can lead to
R. J. Kelly : O. Rixe (*) significant renal toxicities, including hypertension and
National Cancer Institute, Medical Oncology Branch,
proteinuria. This class-effect will be extensively reviewed
Center for Cancer Research,
Bethesda, MD 20892, USA in this article in terms of mechanism, frequency and
e-mail: rixeo@mail.nih.gov clinical implication for the future use of these com-
pounds. In parallel, we will discuss the renal adverse
B. Billemont
events observed with preliminary data obtained with
CERIA (Centre d’Etudes et de Recours sur les Inhibiteurs de
l’Angiogenese), Cochin Hospital, other antiangiogenics blocking a VEGF-independent
Paris, France pathway.
122 Targ Oncol (2009) 4:121–133

Fig. 1 Schematic representation podocyte


of the nephron and the expres- mesangial cell
sion of the targets VGEF

VEGF-R
FGF-R
HIF2 α
PDGF-R β
Dii4

basement membrane HIF1 α

epithelial cell

VGEF

VEGF-R
NOTCH

MET
PDGF-R β

glomerulus
EGF-R
mesangial cell

VEGF-R

EGF-R
MET

capillaries

VEGF-R

Antiangiogenic compounds and renal toxicity VEGF expression in the adult

In the VEGF era, many selective drugs have been brought VEGF is strongly expressed embryologically, playing a
into the clinic. Different strategies have been developed in critical role in new blood vessel formation. In adults, VEGF
two opposite directions: the use of “dirty” drugs with a is a key regulator of many physiologic processes such as
large spectrum of activity contrasting with the develop- wound healing, bone repair and the response of skeletal
ment of very specific compounds, targeting one key muscle to exercise [5]. It also plays a central role in
protein involved in the angiogenic process [3]. This maintaining a normal menstrual and reproductive cycle
duality is not restricted to VEGF-VEGFR inhibition, but with monthly changes occurring in the uterus, ovary and
can be applied to other targeted agents including anti-EGFR breast [6]. VEGF is upregulated in several patho-
molecules [4]. In clinical trials, highly specific or selective physiologic processes (see Table 1). Maharaj et al. have
inhibition of only one of the kinases involved in the proposed a classification involving three levels of expres-
signaling pathways has been associated with sporadic or sion of VEGF in normal tissues. In the “sparse cellular”
limited responses. The selective versus multiple inhibitions expression group, including brain, retina and testes, VEGF
have significant potential implications in terms of activity is expressed primarily by pericytes and vascular stromal
and toxicity. cells [7]. The “intermediate group” of VEGF expression
Targ Oncol (2009) 4:121–133 123

Table 1 Occurrence of proteinuria in clinical trials with bevacizumab

Treatment Cancer No of patients Proteinuria No. of patients (%) Reference

All grades Grade 3–4


B+C BC 229 51 (22.3%) 1 (0.4%) Miller et al. 2005 [85]
B+P BC 365 - 13 (3.5 %) Miller et al. 2007 [86]
B+E BC 38 8 (21%) 0 Dickler et al. 2008 [87]
B + Cy + C BC 46 15 (32%) 1 (0.2%) Dellapasqua et al. 2008 [88]
B all level RCC 76 40 (52.3%) 5 (6.5%) Yang et al. 2003 [89]
3 mg/kg 39 25 (32.5%) 3 (3.9%)
10 mg/kg 37 15 (19.7%) 2 (2.6%)
B + INF RCC 327 59 (18%) 22 (7%) Escudier et al. 2007 [90]
B + INF RCC 366 - 56 (15%) Rini et al. 2008 [91]
B+E RCC 51 (B) - 3 (5.7%) Bukowski et al. 2007 [92]
51 (B + E) - 16 (31%)
B+P+C NSCLC 66 21 (32%) 1 (1.5%) Johnson et al. 2004 [93]
B+P+C NSCLC 434 NA 13 (3.1%) Sandler et al. 2006 [94]
B+E NSCLC 32 9% 0 Herbst et al. 2005 [95]
B + E vs B NSCLC 78 0 0 Herbst et al. 2007 [96]
B + XELOX Vs B + FOLFOX CCR 141 vs 72 - 20 vs 15 Hochster et al. 2008 [97]
B + XELOX vs B + FOLFOX CCR 350 vs 349 4 (<1%) 0 Saltz et al. 2008 [98]
B + XELOX +/- Cetux CCR 192 vs 197 82 (21%) 21 (5%) Tol et al. 2008 [99]
IFL +/- B CCR 393 26.5 % 0.8 % Hurwitz et al. 2004 [100]

B (bevacizumab), C (Capecitabine), P (Carboplatin), E (Erlotinib), Cy (cyclophosphamide), INF (Interferon), Xelox (Capecitabine + oxaliplatin),
FOLFOX (Oxaliplatin + 5FU), Cetux (Cetuximab)

involves cardiac and skeletal muscles. In the “highest VEGFR1 and VEGFR2, and is essential in many physio-
density” group, VEGF is expressed on the epithelia in logic processes. VEGF induces and maintains fenestration.
contact with the fenestrated vasculature of tissues such as Fenestrae on endothelial cells are discontinuities that
the glomerulus, choroid plexus, liver and pancreas. Inter- facilitate movement of particules in and out the circulation
estingly, endothelial cells do not generally express [12].
VEGF [8]. As an exception, aortic endothelial cells
express VEGF, but not the vena cava [9]. The role of Anti-VEGF agents and their associated renal toxicity
expression of VEGF in this large blood vessel is unknown.
VEGF is potentially expressed in every tissue in the adult, Despite its central role in many normal physiologic
and its receptor (VEGFR2) is also expressed in close functions VEGF is also a key regulator of tumor angiogen-
proximity to the VEGF source [10]. esis. This finding has led to the development of several
VEGF-targeted agents that include neutralizing monoclonal
Function of VEGF in the adult antibodies [13] (Fig. 2).

The function of VEGF appears to be pleiotropic. (1) The Bevacizumab


low expression of VEGF seen in the vasculature of tissues
having barrier functions (e.g., retina, brain, testes) may Bevacizumab is an anti-VEGF165 humanized monoclonal
contribute to the impermeability of these vessels [11]. (2) antibody approved by the US Food and Drug Administra-
Inhibition of VEGF leads to an alteration of the microvas- tion for the treatment of metastatic renal cell carcinoma but
culature and in blood vessel regression in a number of also colon, breast and non-small cell lung carcinoma in
normal tissues. (3) VEGF plays an integral role in the combination with chemotherapy. Bevacizumab is specific
normal functioning of fenestrated endothelial cells. These for VEGF (also referred to as VEGF-A) but does not
cells are found in many endocrine tissues as well as in the neutralize other members of the VEGF family (VEGF-B to
kidney, the choroid plexus and the gastrointestinal tract. E) [14]. Monoclonal antibodies, including bevacizumab,
The permeability of endothelial cells is mediated via both have emerged as a class of specific and selective anti-cancer
124 Targ Oncol (2009) 4:121–133

Fig. 2 Site of action of VEGF VEGF-R2 VEGF-R3 PDGF-R HIF1


and VEGFR inhibitors on endo- VEGF-R1 NOTCH
thelial and cancer cells

pericyte C-KIT
endothelial cell
MET
FR
G
VE

tumor cell
Raf

HIF MEK

Erk

PDGF-R
tumor
VEGF-R

molecules [15]. The mechanism of action of bevacizumab The occurrence of renal toxicities demonstrates the
has been extensively studied [16]. The current view of its absence of total selectivity for tumor cell targeting and
angiogenesis inhibition involves a complex series of events has been extensively documented by Eremina et al. [22].
including the classic sprouting angiogenesis inhibition Putative causes of nephrotoxicity have been suggested
(including inhibition of new blood vessel growth and including an indirect effect of the therapy (called “off-
vascular regression), modification of vessel permeability target” toxicity related to immunologic disorders related to
and vascular constriction, direct effect on tumor cell the monoclonal antibody) or “on-target” effects due to the
function, offsetting of effects of chemotherapy induction co-expression of VEGF on the normal kidney. The role of
of VEGF levels, but also bone marrow-derived endothelial other co-morbidities and drug interactions need to be
progenitor cells inhibition. Induction of endothelial cell further identified to determine the individual patient
apoptosis is one the major effect of bevacizumab. VEGF susceptibility to bevacizumab toxicity.
plays a significant role in numerous pro-survival pathways In a very elegant publication, Eremina et al. [23]
in endothelial cell homeostasis [17] including activation of reported six cases of patients with proteinuria associated
BCl-2, Akt and survivin. with thrombotic microangiopathy (TMA) after bevacizu-
The safety profile of bevacizumab has been studied in mab administration. From this clinical evidence, the authors
randomized control clinical trials. Clinically relevant side developed an animal model to document the mechanism of
effects relating to bevacizumab include hypertension, renal toxicity. Using a knock-out mice model, local genetic
proteinuria, arterial thromboembolic events, wound healing ablation of VEGF production in the kidney mimics the
complications and gastro-intestinal perforation [18]. TMA observed in humans. In this study, a lack of VEGF
One of the most common is proteinuria, which is production by podocytes was obtained and mice developed
reported in 21% to 64% of patients. Nephrotic-range proteinuria after a period of 4 weeks followed by the
proteinuria denotes structural damage to the glomerular occurrence of hypertension. The kidneys of all the KO mice
filtration barrier and occurs in 1% to 2% of bevacizumab- were pale and shrunken, while electron microscopy of the
treated patients [19]. Acute renal deficiency or creatinine glomeruli displayed typical features of TMA. When disease
elevation was rarely reported [20]. More recently, several progressed the following alterations were observed: (1)
cases of thrombotic micro-angiopathy have been docu- altered podocytes morphology, (2) swollen endothelial cells
mented. This condition is associated with arterial hyperten- and (3) alteration of fenestrated endothelium. Interestingly,
sion, acute renal deficiency, proteinuria and hematuria [21]. schistocytes were found in blood smears from more than
Targ Oncol (2009) 4:121–133 125

50% of the animals. These findings are similar with those [29]. Similarly to bevacizumab, 2 cases of thrombotic
reported in humans. They suggest a critical role for micro-angiopathy were also reported [30].
VEGF within the glomerulus. No involvement of the
peripheral microvasculature was reported and extra-renal Sorafenib
circulating VEGF levels were not affected in this KO
mice model. The authors conclude that renal damage Sorafenib is a tyrosine kinase inhibitor of VEGF-R, PDGF-
induced by bevacizumab is a direct reduction of R, C-kit and BRAF. In a phase II study conducted for the
glomerular VEGF production, or as previously described treatment of hepatocellular carcinoma, proteinuria was
an “on-target” effect of the drug. It appears that reported in 19 patients (41%) [31]. These results were not
Bevacizumab administration is associated with damage replicated in the phase III study, as hypophosphastemia was
to the glomerular endothelium suggesting that local the only renal toxicity reported [32].
VEGF-A production is required for the maintenance of Grade 1 creatinine elevation was reported in a phase II
the integrity of this specialized vascular bed and ulti- study in prostatic patients treated with sorafenib [33]. In
mately there is interference with the critical cross-talk addition, a similar study performed in 22 androgen-
between podocytes and endothelium in the glomerulus. A independent prostate cancer patients revealed 3 cases of
similar observation has been reported with preeclampsia, a hypocalcemia, 2 cases of hyponatremia and 2 cases of
pregnancy-specific disorder that complicates approximate- hypophosphastasemia [34]. No significant renal toxicities
ly 5% of all pregnancies, making it perhaps the most were reported in the large phase III TARGET study
common glomerular disease in the world. It is character- conducted in metastatic renal cell carcinoma [35]. Izzedine
ized by new-onset hypertension and proteinuria, second- et al. recently reported interstital nephritis associated with
ary to the previously described bevacizumab induced skin toxicity [36]. The combination of sorafenib and
alterations that occur in the glomerulus. Recent evidence bevacizumab was associated with proteinuria and hyper-
suggests that these alterations in the glomerulus are tension and was reported as the dose limiting toxicity in a
mediated by an extra-renal overproduction of a soluble recently published phase 1 study [37].
VEGF receptor that denies normal functioning glomerular
endothelial cells of VEGF. This subsequently leads to VEGF-independent inhibition
cellular injury and disruption of the filtration apparatus
with subsequent proteinuria [24]. HIF-1alpha inhibition
Drug induced hypertension is most likely secondary to
the renal toxicity of bevacizumab and it is a clinical Hypoxic-inductible factors (HIFs) are heterodimeric basic
symptom associated with thrombotic micro-angiopathy helix-loop-helix/PAS proteins that induce the transcription
[25]. In addition, VEGF starvation reduces endothelial of a diverse array of genes to affect the hypoxic response.
nitric oxide production, which increases blood pressure by HIFs are composed of an α subunit (HIF-1α, HIF-2 α and
causing vasoconstriction. Hypertension can also be second- HIF-3 α) and a β subunit (HIF-1β or ARNT) [38]. There
ary to kidney injury or vascular rarefaction, resulting in an are multiple splice variants of HIF-1 α and HIF-3 α, with
increase of systemic resistance [26]. These observations dominant negative proteins being produced in some tissues.
strongly support the absence of absolute tumor selectivity HIFs are key elements required to mediate cellular
of anti-VEGF agents, emphasizing the critical role that adaptation to low O2 by activating the transcription of
VEGF plays in normal tissues homeostasis. target genes involved in angiogenesis, metabolism, and
extra-cellular matrix remodeling [39]. Overexpression of
Sunitinib HIFs promotes tumorigenesis, but paradoxically, there is
genetic evidence for a tumor suppressor role for HIF-1α.
Sunitinib is an oral multikinase inhibitor which targets The critical role of HIFs in the development of renal cell
VEGF-R, PDGF-R, and c-kit [27]. In the initial phase I carcinoma is demonstrated by an increase in HIF-2α
study, Faivre et al. reported a case of grade 3 hypertension, expression via c-Myc activation in sporadic VHL-deficient
associated with a paradoxical 30-fold increase circulating clear cell RCC and is the basis for the current development
VEGF level [28]. In a phase II study conducted in renal cell of anti-HIF-1α targeted therapies in this setting.
carcinoma, creatinine elevation was present in 14% of the In human tissues, HIF-1α messenger RNA expression is
63 patients. In addition, grade 2 (2 patients, 3.2%) and generally ubiquitous with high expression in the kidneys
grade 3 (1 patient, 1.6%) hypertension were respectively (predominantly in the cortical and medullary ducts, and
reported in this analysis. In the large phase III study (renal glomerular cells) [40]. The expression of HIF-2α is
cell carcinoma), elevation of creatinine level was reported detectable in podocytes, as well as in cortical and medullary
in 66 patients (17.6 %) and grade 3 (0.3%) in one patient endothelial and interstitial cells. HIF-2α may play a role in
126 Targ Oncol (2009) 4:121–133

the development of the tubular system, but the real role of Similarly, the notch signaling pathway is overexpressed in
HIFs in nephrogenesis needs to be further delineated. the proximal tubules and is involved during acute injury
Chronic hypoxia has been described as the final common such as tubular necrosis. It is clear that stimulation of the
pathway to end-stage renal failure, and HIFs are seen as the notch pathway in the normal healthy adult kidneys can lead
master switch of the hypoxic adaptation response. HIFs to complications. Further studies are required to determine
often have contrasting effects on the kidneys, from the if notch activation is either a response mechanism or a
beneficial cytoprotective effect seen in acute ischemia- primary cause of the renal disease.
reperfusion injury to a detrimental role in inflammation At present inhibition of the notch pathway for the
with a profibrotic effect in chronic renal hypoxia. treatment of cancer is still conceptual. Further studies are
Recently, the first in human phase I study with an anti- required to determine the exact role notch plays in terms of
HIF1α RNA antagonist has been reported. EZN-2968 is a nephroprotection or nephrotoxicity. Future notch receptor
third generation oligonucleotide (locked nucleic acid). In inhibitions will also require a high level of specificity as
vivo, this compound has a broad distribution into major different notch receptors are responsible for different
organs including the kidneys [41]. No renal toxicities have functions with notch2, but not notch1, being required for
been reported in mice. In the phase I study conducted in 18 fate acquisition in the mammalian nephron [49].
patients, one patient experienced grade 3 hypertension.
Interestingly, no proteinuria or renal dysfunction has been
reported to this point [42]. Epidermal growth factor inhibition and renal toxicity

Notch inhibition The epidermal growth factor receptor (EGFR) is part of a


family of four tyrosine kinases whose aberrant activity
The notch signaling pathway plays a pivotal role in plays a key role in growth and metastasis of solid tumors:
angiogenesis but appears to be both independent of and epidermal growth factor receptor (EGFR; erbB-1), erbB-2
run parallel to the well described VEGF/VEGFR system (Her2 or neu), erbB-3, and erbB-4 [50]. There are six direct
[43]. Notch receptors comprise a family of transmembrane binding ligands known for EGFR, including EGF, trans-
receptors, their ligands and the subsequent transcription forming growth factor, amphiregulin, betacellulin, epiregu-
factors. They are expressed by various cell types and are lin, and heparin-binding EGF. Ligand binding to the
involved in cell differentiation, cell fate, and cell prolifer- extracellular domain of EGFR leads to receptor dimeriza-
ation. The four membrane receptors (notch 1, 2, 3 and 4) tion, causing the activation of downstream signaling path-
interact with transmembrane ligands (e.g., jagged 1, Dll4). ways by activation of the cytosolic kinase domain of the
Dll4 is expressed exclusively by endothelial cells and receptor tyrosine kinase and cross-autophosphorylation of
represents a major stimulator (via its notch receptor) of the receptors [51].
angiogenesis [44]. Knockout of only Dll4 in mice is lethal to It has also been shown that erbB-2 is the preferential
the embryo and is seen as an essential signal for normal heterodimerization partner of EGFR compared with the rest
vascular development. The notch-Dll4 complex is upregu- of the erbB family members [52]. At least three main
lated in tumor vasculature and drugs targeting this pathway pathways of EGFR signaling supporting cancer develop-
are in early phases of preclinical development. Studies ment and progression can be identified. They include (1)
involving neutralizing antibodies against Dll4, and inhibitors phosphatidylinositol 3-kinase (PI3K) leading to Akt activa-
of the notch receptor are ongoing. Preclinical data suggest tion and suppression of apoptosis, (2) Grb2 and Sos leading
that drugs that target the Dll4-ligand can paradoxically lead to the activation of p21ras driving the cell cycle, and (3)
to an increase in immature tumor vasculature, however, these phospholipase C-γ1 phosphorylation leading to PIP2-
new vessels are unable to support tumor growth [45]. related actin reorganization.
Podocytes are a type of epithelial glomerular cell Numerous anti-EGFR agents have been developed and
involved in the ultrafiltration process that occurs in the are used routinely in clinical practice for the treatment of
kidney [46]. Alteration of several podocyte proteins has several solid tumors including colon cancer, lung cancer
previously been described in the development of glomer- and head and neck tumors. Routinely used drugs include
ulosclerosis. The notch pathway plays a crucial role in renal small molecule tyrosine kinase inhibitors (e.g., erlotinib,
development during embryogenesis [47]. There is however gefitinib) or monoclonal antibodies (e.g., cetuximab,
little active notch1 detected in mature adult kidneys under panitinumab). HER2 (erbB-2) overexpression is associated
normal conditions. Activation of the notch pathway in with an aggressive phenotype in breast cancer. Trastuzumab
mature podocytes has been associated with glomerular and lapatinib have successfully been developed for the
disease, and pharmacological inhibition of notch cleavage treatment of metastatic and locally advanced breast cancer
can prevent podocyte apoptosis and albuminuria [48]. overexpressing HER2.
Targ Oncol (2009) 4:121–133 127

In the kidney, EGFR is expressed mainly in distal, published case reports of gefitinib-induced renal toxicities
collecting, and proximal tubules. It has also been detected have highlighted a incidences of nephrotic syndrome and
in glomerular capillary walls and other vascular compo- tubulointerstitial nephritis after the administration of gefi-
nents (e.g., mesengial and parietal epithelial cells, peritub- tinib. In the first case, mild infiltration of lymphocytes have
ular capillaries and arterioles). Functionally, EGFR has a led to sclerotic changes in renal glomeruli and suggest an
dual role in renal epithelia [53]: (1) reactive species- immune reaction to gefitinib [57]. In the second case, after
dependent activation of EGFR leads to cell death in renal a two-year period of gefitinib administration, immunofluo-
proximal tubule cells exposed to cisplatin, and (2) func- rescence revealed a predominance of mesangial IgA and C3
tional inactivation of EGFR in renal proximal tubular cells deposits, suggesting an IgA nephropathy [58]. It has been
reduces tubular-interstitial lesions after renal injury [54]. suggested that long-term administration of gefitinib could
Once activated the EGFR/Ras/MEK/ERK pathway serves a inhibit the normal turnover of tubular epithelial cells,
pro-survival role in renal epithelia under moderate oxida- leading to interstitial damage [59].
tive stress. Severe oxidative stress induced by ischemic Cetuximab, a monoclonal antibody against the extracel-
conditions can result in necrotic death via a mechanism that lular domain of the EGFR, has been approved for the
disconnects EGFR from ERK activation in both renal treatment of metastatic colon carcinoma in combination
proximal and distal cells. These observations explain the with irinotecan, and more recently for the treatment of
occurrence of renal toxicities after the use of EGFR locally advanced head and neck carcinoma in combination
modulators in humans. However, EGFR expression level with radiation therapy [60]. No renal toxicity, including
is lower in normal epithelial tissues in comparison with proteinuria, hypertension or renal dysfunction, has been
cancer cells. HER2 is commonly expressed in fetal reported with this agent. In a recent survey conducted by
epithelial cells of the human kidney and to a lesser the EORTC in patients with head and neck carcinoma
degree in normal human adult kidneys. HER2 is treated with cetuximab, the authors do not report renal
implicated in the collecting tubular cyst formation and dysfunction [61].
enlargement in polycystic kidney disease in concert with
EGFR. HER2 inhibitors

EGFR inhibitors Trastuzumab is a recombinant monoclonal antibody


directed against the extracellular domain of the HER2
Erlotinib is a commonly used epidermal growth factor protein [62]. Data from phase I to III clinical trials and from
receptor (HER1/EGFR) tyrosine kinase inhibitor [55]. It post marketing surveillance has not highlighted significant
inhibits ATP binding to HER1/EGFR tyrosine kinase in renal toxicities [63]. Trastuzumab is however teratogenic
normal and tumor cells. The tyrosine kinase of HER1/ when administered during pregnancy with normal fetal
EGFR is a target for therapeutic intervention in many development of the kidneys being impaired [64].
malignancies including ovarian, head, neck, lung, breast, Lapatinib is a tyrosine kinase inhibitor approved for use
bladder and other squamous cell cancers. The most in combination with capecitabine to treat advanced or
common side effects in patients receiving erlotinib are skin metastatic breast cancers overexpressing HER2. Lapatinib
rash, fatigue and diarrhea. Other reported side-effects is a dual inhibitor of EGFR and HER2, and no renal
included interstitial lung disease. No renal toxicity has adverse events have been reported in the 44 published
been described with this agent. In the phase I and phase II clinical studies [65].
studies conducted in combination with bevacizumab, Other HER2 inhibitors (pertuzumab, ertumaxomab and
hypertension, proteinuria and renal dysfunction have been trastuzumab-MCC-DM1) are in early phases of develop-
reported, but do not significantly differ between the ment but no nephrotoxicity has as yet been reported.
combination arm and bevacizumab arm. Miller et al. [56]
recently reported that patients with renal dysfunction can
tolerate a full dose of erlotinib (150 mg daily) and seem to Other targeted therapies and renal toxicity
have an erlotinib clearance similar to patients without organ
dysfunction. Imatinib is a tyrosine kinase inhibitor that targets the
Gefitinib is similar to erlotinib and is also an orally Abelson (Abl) tyrosine kinase with high selectivity.
active small-molecule tyrosine kinase inhibitor of EGFR. Imatinib but also inhibits CD117 (c-Kit) and platelet-
Preclinical data from the phase I study of gefitinib did not derived growth factor (PDGF)-α and PDGF-β at concen-
indicate any clinically significant changes in renal param- trations of approximately 0.5 µmol/l in vitro. PDGF-β and
eters. The toxicity profile is similar to erlotinib, including c-kit are both expressed in the kidney. PDGF-β expression
diarrhea, rash, acne, dry skin, nausea, and vomiting. Two has been reported in proximal tubes, mesangial and
128 Targ Oncol (2009) 4:121–133

interstitial cells [66]. c-Kit is also expressed in both Interestingly, no renal toxicities have been reported so far
proximal and distal tubules. In the initial large study with the anti-MET antibodies. However, as Met regulates
conducted by Druker et al in chronic myeloid leukemia, and mediates maintenance of the normal adult collecting
renal failure was reported in less than 1% of the patients duct, putative renal toxicities of MET inhibitors are
[67]. When a higher dose (800 mg daily) was tested in expected.
patients with renal cell carcinoma, the occurrence of grade Newer agents involving molecular targeted therapies
II renal toxicity was higher (14%) [68]. In addition, several affecting many key novel pathways—HDAC (histone
cases of acute tubular necrosis were reported and docu- deacetlyase inhibitors), HSP90 (heat shock protein 90
mented with renal biopsies. Francois recently reported a inhibitors), PARP (Poly (ADP-ribose) polymerase-enzyme
case of modest renal impairment associated with a Fanconi inhibitors), Hegdehog, IGFR, PI3k, AKT, and Ras inhib-
syndrome [69]. Electronic microscopy showed vacuoles in itors—are currently under pre-clinical or clinical develop-
some tubular cells, suggesting proximal tubular injury. ment. No specific renal toxicities have been reported as yet
Imatinib induced tubular necrosis has also been reported by but detailed safety analysis will be required as many of
Foringer and Pou [70], necessitating hemodialysis. In these cellular pathways represent important signals for renal
addition, one case of thrombotic microangiopathy (TMA) cell homeostasis.
with renal and extra-renal expression has been reported,
requiring definitive hemodyalisis [71]. Patients with preex-
isting renal dysfunction should be monitored closely for the Recommendations for the management of renal
development of renal failure and tubular toxicity when toxicities
taking imatinib.
Nilotinib is a second-generation imatinib analog with a Renal impairment can be assessed by measuring both
similar spectrum of activity. It is at an earlier phase of its glomerular and tubular functions in the kidney. The
development than imatinib but as yet no renal toxicity has measurement of creatinine clearance reflects the glomerular
been reported [72]. filtration rate. In daily clinical practice, renal function is
Dasatinib is the first of the second-line ABL inhibitors estimated by the Cockroft formula (Cockcroft, Gault 1976)
approved by the Food and Drug Administration. This is a and more accurately by the MDRD (Modification of Diet in
multi-kinase inhibitor that targets SRC (a family of proto- Renal Disease) formula [77]. Urinary albumin excretion
oncogenic tyrosine kinases) and many other kinases besides (determined by urine dipstick testing, spot urine sampling,
BCR-ABL [73]. No renal toxicity has been currently urinary protein/creatinine ratio (UPC) or 24-hour protein
published. determination) explores the integrity of the glomeruli.
MET tyrosine kinase is a cell surface receptor for Tubular function is assessed by the excretion of glucose,
hepatocyte growth factor (HGF). HGF conveys a combina- pH, osmolarity, urinary uric acid, calcium, phosphorus and
tion of pro-migratory, mitogenic and anti-apoptotic signals magnesium. Based on these parameters, a grading system
[74]. MET is a candidate for targeted therapeutic interven- of renal toxicities has been established and is available in
tion. Several genetic alterations including translocation, the National Cancer Institute Common Terminology Crite-
amplification or activating mutations have been reported in ria document. This classification focuses on an alteration of
several types of malignancies. Germline MET gene the glomerular filtration rate and metabolic disorders (Table 2).
mutations and somatic MET gene mutations are the key Patients with cancer have an increased risk of developing
molecular events described respectively in hereditary renal failure [78]. A detailed history evaluating specific co-
papillary renal cell carcinoma and in its sporadic form. morbidities (e.g., diabetes, hypertension, nephrectomy)
HGF (and the ensuing activation of MET) plays a critical needs to be performed at baseline. In some cases, cancer
role in the control of tissue homeostasis (including the may cause obstruction in the urinary tract or lead to a direct
normal kidney) but also in several degenerative diseases infringement of the renal parenchyma. Tumors may also be
including progressive nephropathies [75]. Several strategies associated with metabolic abnormalities (e.g., hypercalce-
have been pursued to inhibit MET. HGF or MET mia, hepato-renal syndrome) [79]. Finally, the prior use of a
neutralizing antibodies and small-molecule MET inhibitors conventional first line anti-cancer agent (e,g,, platinum
are currently under investigation in phase I and II studies. salts) can lead to nephrotoxicity and impaired kidney
XL880 clinical trials began in 2005 and phase II trials are function even before initiating a second or third line
currently in process. In the papillary RCC phase II study, a targeted therapy.
15% (including 8% grade 3) rate of hypertension (mainly Nephrotoxic risk must be evaluated before starting any
associated with proteinuria) has been reported [76]. This anti-angiogenic treatment. This includes a thorough history,
can be related to the MET inhibition, and also to the clinical examination and laboratory investigations evaluat-
additional tyrosine kinases inhibition including VEGFR2. ing for renal dysfunction. Following the initiation of
Targ Oncol (2009) 4:121–133 129

Table 2 Grading for renal toxicities

Grade Creatinine Proteinuria Glomerular filtration rate

1 Normal −1.5×ULN 1+or 0.15–1.0 g / 24 h <75–50% LLN


2 1.5–3.0×ULN 2+to 3+or >1.0–3.5 g/24 h <50–25% LLN
3 3.0–6.0×ULN 4+or >3.5 g/24 h <25% LLN, chronic dialysis non indicated.
4 6.0×ULN Nephrotic syndrome Chronic dialysis (or renal transplant) indicated.

ULN: Upper Limit Normal LLN: Lower Inferior Normal

treatment patients should be monitored every two weeks blood pressure control is less than optimal, the patient
both clinically and with bioassays and urine testing. In the should be referred to a nephrologist. Standard oral
presence of abnormal blood and urine values or signs antihypertensive agents used to manage antiangiogenic-
suggestive of thrombotic micro-angiopathy, an expert related hypertension include angiotensin-converting en-
nephrological opinion should be sought (Table 3). zyme (ACE) inhibitors, beta blockers, calcium channel
Blood pressure and proteinuria must be monitored blockers, and diuretics. The presence of hypertension and
before, during and after this medical treatment, even in a proteinuria would favor the use of ACE inhibitors or
palliative setting. Intervention must be both timely and angiotensin receptor blockers, due to their renal protective
aggressive to avoid consequences of acute hypertension. effect with the restoration of nephrin expression. Calcium
No clear consensus for the choice of anti-hypertensive channel blockers may interact with small molecules through
agents has been established. In the presence of hyperten- cytochrome CYP3A4 and could expose patients to a risk of
sion, careful monitoring of blood pressure is recommended. underdosage or overdosage of the antiangiogenic molecules.
Patients during therapy should be advised to purchase a
home blood pressure monitoring system. Blood pressure
should be checked at home at least once daily (in the Conclusion
morning), and patients should be advised to contact the
clinic if they note elevated blood pressure levels. Analysis of the literature reveals that significant renal
Antiangiogenic-related hypertension should be treated with toxicities secondary to targeted molecular therapies exist.
oral antihypertensive therapy. After a titration for effect, if This phenomenon underlines the absence of selectivity of

Table 3 Summary of renal tox-


icities associated with molecular Class Symptoms Nephron damage (biopsy proven)
targeted therapies
ANTIANGIOGENICS
VEGF inhibitors Hypertension/Proteinuria Glomerulonephritis
TMA
VEGFR inhibitors Hypertension/Proteinuria Glomerulonephritis
Interstitial Nephritis
HIF inhibitors NR
Notch inhibitors ND
OTHERS
HER inhibitors
EGFR inhibitors Proteinuria Acute Tubular Necrosis
IgA nephropathy
HER2 inhibitors NR NR
C-KIT inhibitors Hypertension/Proteinuria TMA
Acute Tubular Necrosis
MET inhibitors Hypertension/Proteinuria ND
HSP90 inhibitors NR
NR: not reported PARP inhibitors NR
ND: no data available IGF1R inhibitors NR
TMA: thrombotic PI3-kinase inhibitors NR
microangiopathy
130 Targ Oncol (2009) 4:121–133

these molecules, mainly with regards to agents that are especially with antiangiogenics, renal function must be
classed as antiangiogenics. Unfortunately many of the so- assessed by calculation of creatinine clearance even when
called targeted agents not only affect markers that are serum creatinine is within the normal range [78].
expressed in tumor cells but also those expressed in normal
tissues, specifically in the kidney. In a recent analysis of Acknowledgements We thank the Fondation Martine Midy, Paris,
France for technical assistance in preparation of this manuscript.
kinase inhibitors, Karaman et al. [80] reported a wide
diversity of interactive patterns which could explain the Conflict of interest statement No funds were received in support of
occurrence of renal adverse events. this study, nor were any benefits received from a commercial party
Most of the targets modulated by anti-cancer molecular related directly or indirectly to the subject of this manuscript, except
those directed solely to a research fund, foundation, educational
therapies are expressed on the normal nephron. Based on institution or other non-profit organization with which one or more of
the current published data, we can identify three different the author(s) is/are associated.
categories of nephrotoxic agents: (1) high risk of nephrop-
athy, in terms of frequency and severity, mainly represented
by the anti-VEGF/VEGFR agents and other anti- References
angiogenics, (2) intermediate risk of nephropathy, with the
c-kit inhibitors, and (3) low-risk, including anti-EGFRs. 1. Dvorak HF (2002) Vascular permeability factor/vascular endo-
Post marketing surveillance data in a large unselected thelial growth factor: a critical cytokine in tumor angiogenesis
patient population will however allow us to evaluate and a potential target for diagnosis and therapy. Clin Oncol 20
(21):4368–4380
whether the risks of drug induced nephrotoxicity have been 2. Ellis LM, Hicklin DJ (2008) Pathways mediating resistance to
underestimated in phase I/II/III clinical trials. vascular endothelial growth factor-targeted therapy. Clin Cancer
Studies exploring the mechanisms of toxicities are Res 14(20):6371–6375
limited, but in order to properly manage hypertension and 3. Lorusso PM, Eder JP (2008) Therapeutic potential of novel
selective-spectrum kinase inhibitors in oncology. Expert Opin
proteinuria we will require an improved understanding of Investig Drugs 17(7):1013–1028
the pathogenesis of drug induced nephrotoxicities. Mecha- 4. Raymond E, Faivre S, Armand JP (2000) Epidermal growth
nisms causing renal damage are various and class depen- factor receptor tyrosine kinase as a target for anticancer therapy.
dant. Toxicities with antiangiogenics are now well Drugs 60(Suppl 1):15–23
5. Li J, Hampton T, Morgan JP, Simons M (1997) Stretch-induced
identified and have been documented with preclinical VEGF expression in the heart. J Clin Invest 100(1):18–24
animal models and data obtained from several renal 6. Maharaj AS, Saint-Geniez M, Maldonado AE, D’Amore PA
biopsies. Clinical investigators and medical oncologists in (2006) Vascular endothelial growth factor localization in the
general might consider performing a renal biopsy more adult. Am J Pathol 168(2):639–648
7. Maharaj AS, D’Amore PA (2007) Roles for VEGF in the adult.
frequently when a nephropathy related to targeted therapies Microvasc Res 74(2–3):100–113
is suspected, especially in the earlier phases of develop- 8. Couffinhal T, Kearney M, Witzenbichler B, Chen D, Murohara
ment. Hypertension and proteinuria are the most common T, Losordo DW, Symes J, Isner JM (1997) Vascular endothelial
side effects of antiangiogenic therapy, including VEGF and growth factor/vascular permeability factor (VEGF/VPF) in
normal and atherosclerotic human arteries. Am J Pathol 150
VEGFRs inhibitors. (5):1673–1685
Renal toxicities observed after antiangiogenic treatments 9. Stalmans I, Ng YS, Rohan R, Fruttiger M, Bouché A, Yuce A,
do not seem to be dose-dependant or dose-cumulative. For Fujisawa H, Hermans B, Shani M, Jansen S, Hicklin D,
instance, VEGF-trap renal toxicity has been observed at a Anderson DJ, Gardiner T, Hammes HP, Moons L, Dewerchin
M, Collen D, Carmeliet P, D’Amore PA (2002) Arteriolar and
low dose in a phase I study [81], but similar effects were venular patterning in retinas of mice selectively expressing
not seen at higher doses. These toxicities are usually VEGF isoforms. J Clin Invest 109(3):327–336
reversible and offer the possibility of further therapies. 10. Monacci WT, Merrill MJ, Oldfield EH (1993) Expression of
However, the reintroduction of a compound of the same vascular permeability factor/vascular endothelial growth
factor in normal rat tissues. Am J Physiol 264(4 Pt 1):
class should be carefully evaluated in the absence of robust C995–C1002
data and greater experience. 11. Kilic U, Kilic E, Järve A, Guo Z, Spudich A, Bieber K, Barzena
Hypertension is a potential indirect marker of anti- U, Bassetti CL, Marti HH, Hermann DM (2006) Human vascular
angiogenic induced renal toxicity. Increased blood pressure endothelial growth factor protects axotomized retinal ganglion
cells in vivo by activating ERK-1/2 and Akt pathways. J
during a-VEGF treatment (including bevacizumab [82], Neurosci 26(48):12439–12446
sunitinib [83] and axitinib [84]) is associated with clinical 12. Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS,
efficacy. The direct relationship between renal toxicity and Dvorak HF (1983) Tumor cells secrete a vascular permeability
efficacy needs to be addressed in future clinical trials. factor that promotes accumulation of ascites fluid. Science. 219
(4587):983–985
Finally, we have to remember that the frequency of 13. Krämer I, Lipp HP (2007) Bevacizumab, a humanized anti-
chronic renal insufficiency among cancer patients remains angiogenic monoclonal antibody for the treatment of colorectal
high. Before the initiation of an anticancer treatment, cancer. J Clin Pharm Ther 32(1):1–14
Targ Oncol (2009) 4:121–133 131

14. Presta LG, Chen H, O’Connor SJ, Chisholm V, Meng YG, microangiopathy secondary to VEGF pathway inhibition by
Krummen L, Winkler M, Ferrara N (1997) Humanization of an sunitinib. Nephrol Dial Transplant 24(2):682–685
anti-vascular endothelial growth factor monoclonal antibody for 31. Abou-Alfa GK, Schwartz L, Ricci S, Amadori D, Santoro A,
the therapy of solid tumors and other disorders. Cancer Res 57 Figer A, De Greve J, Douillard JY, Lathia C, Schwartz B, Taylor
(20):4593–4599 I, Moscovici M, Saltz LB (2006) Phase II study of sorafenib in
15. Tassev DV, Cheung NK (2009) Monoclonal antibody therapies patients with advanced hepatocellular carcinoma. J Clin Oncol
for solid tumors. Expert Opin Biol Ther 9(3):341–353 24(26):4293–4300
16. Ferrara N, Hillan KJ, Gerber HP, Novotny W (2004) Discovery 32. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF,
and development of bevacizumab, an anti-VEGF antibody for de Oliveira AC, Santoro A, Raoul JL, Forner A, Schwartz M,
treating cancer. Nat Rev Drug Discov 3(5):391–400 Porta C, Zeuzem S, Bolondi L, Greten TF, Galle PR, Seitz JF,
17. Dvorak HF (2002) Vascular permeability factor/vascular endo- Borbath I, Häussinger D, Giannaris T, Shan M, Moscovici M,
thelial growth factor: a critical cytokine in tumor angiogenesis Voliotis D, Bruix J, SHARP Investigators Study Group (2008)
and a potential target for diagnosis and therapy. J Clin Oncol 20 Sorafenib in advanced hepatocellular carcinoma. N Engl J Med
(21):4368–4380 359(4):378–390
18. Gordon MS, Margolin K, Talpaz M, Sledge GW Jr, Holmgren E, 33. Chi KN, Ellard SL, Hotte SJ, Czaykowski P, Moore M, Ruether
Benjamin R, Stalter S, Shak S, Adelman D (2001) Phase I safety JD, Schell AJ, Taylor S, Hansen C, Gauthier I, Walsh W,
and pharmacokinetic study of recombinant human anti-vascular Seymour L (2008) A phase II study of sorafenib in patients with
endothelial growth factor in patients with advanced cancer. J chemo-naive castration-resistant prostate cancer. Ann Oncol 19
Clin Oncol 19(3):843–850 (4):746–751
19. Frangié C, Lefaucheur C, Medioni J, Jacquot C, Hill GS, Nochy 34. Dahut WL, Scripture C, Posadas E, Jain L, Gulley JL, Arlen PM,
D (2007) Renal thrombotic microangiopathy caused by anti- Wright JJ, Yu Y, Cao L, Steinberg SM, Aragon-Ching JB, Venitz
VEGF-antibody treatment for metastatic renal-cell carcinoma. J, Jones E, Chen CC, Figg WD (2008) A phase II clinical trial of
Lancet Oncol 8(2):177–178 sorafenib in androgen-independent prostate cancer. Clin Cancer
20. Roncone D, Satoskar A, Nadasdy T, Monk JP, Rovin BH Res 14(1):209–214
(2007) Proteinuria in a patient receiving anti-VEGF therapy for 35. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels
metastatic renal cell carcinoma. Nat Clin Pract Nephrol 3 M, Negrier S, Chevreau C, Solska E, Desai AA, Rolland F,
(5):287–293 Demkow T, Hutson TE, Gore M, Freeman S, Schwartz B, Shan
21. Zhu X, Wu S, Dahut WL, Parikh CR (2007) Risks of proteinuria M, Simantov R, Bukowski RM, TARGET Study Group (2007)
and hypertension with bevacizumab, an antibody against Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J
vascular endothelial growth factor: systematic review and meta- Med 356(2):125–134
analysis. Am J Kidney Dis 49(2):186–193 36. Izzedine H, Brocheriou I, Rixe O, Deray G (2007) Interstitial
22. Eremina V, Baelde HJ, Quaggin SE (2007) Role of the VEGF—a nephritis in a patient taking sorafenib. Nephrol Dial Transplant
signaling pathway in the glomerulus: evidence for crosstalk 22(8):2411
between components of the glomerular filtration barrier. Nephron 37. Azad NS, Posadas EM, Kwitkowski VE, Steinberg SM, Jain L,
Physiol 106(2):32–37 Annunziata CM, Minasian L, Sarosy G, Kotz HL, Premkumar A,
23. Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M, Cao L, McNally D, Chow C, Chen HX, Wright JJ, Figg WD,
Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH, Kohn EC (2008) Combination targeted therapy with sorafenib
Gerber HP, Ferrara N, Barisoni L, Alpers CE, Quaggin SE and bevacizumab results in enhanced toxicity and antitumor
(2008) VEGF inhibition and renal thrombotic microangiopathy. activity. J Clin Oncol 26(22):3709–3714
N Engl J Med 358(11):1129–1136 38. Haase VH (2006) Hypoxia-inducible factors in the kidney. Am J
24. Levine RJ, Lam C, Qian C, Yu KF, Maynard SE, Sachs BP, Sibai Physiol Renal Physiol 291(2):F271–F281
BM, Epstein FH, Romero R, Thadhani R, Karumanchi SA 39. Schofield CJ, Ratcliffe PJ (2004) Oxygen sensing by HIF
(2006) CPEP study group. Soluble endoglin and other circulating hydroxylases. Nat Rev Mol Cell Biol 5(5):343–354
antiangiogenic factors in preeclampsia. N Engl J Med 355 40. Bernhardt WM, Schmitt R, Rosenberger C, Münchenhagen PM,
(10):992–1005 Gröne HJ, Frei U, Warnecke C, Bachmann S, Wiesener MS,
25. George BA, Zhou XJ, Toto R (2007) Nephrotic syndrome after Willam C, Eckardt KU (2006) Expression of hypoxia-inducible
bevacizumab: case report and literature review. Am J Kidney Dis transcription factors in developing human and rat kidneys.
49(2):e23–e29 Kidney Int 69(1):114–122
26. Hoenig MR, Bianchi C, Rosenzweig A, Sellke FW (2008) The 41. Greenberger LM, Horak ID, Filpula D, Sapra P, Westergaard M,
cardiac microvasculature in hypertension, cardiac hypertrophy Frydenlund HF, Albaek C, Schroder H, Orum H (2008) A RNA
and diastolic heart failure. Curr Vasc Pharmacol 6(4):292–300 antagonist of hypoxia-inducible factor-1alpha, EZN-2968, inhib-
27. Izzedine H, Billemont B, Thibault F, Rixe O (2007) New its tumor cell growth. Mol Cancer Ther 7(11):3598–3608
challenges in kidney cancer therapy: sunitinib. Ann Oncol 18 42. Lewis N, Cohen RB, Nishida Y et al (2008) Phase I, pharmaco-
(Suppl 9):ix83–ix86 kinetic (PK), dose-escalation study of EZN 2968, a novel
28. Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N, hypoxia-inducible factor-1alpha RNA antagonist, administered
Bello C, Deprimo S, Brega N, Massimini G, Armand JP, Scigalla weekly in patients with solid tumors. 20th EORTC-NCI-AACR
P, Raymond E (2006) Safety, pharmacokinetic, and antitumor Symposium on Molecular Targets and Cancer Therapeutics,
activity of SU11248, a novel oral multitarget tyrosine kinase Geneva: abstract #398
inhibitor, in patients with cancer. J Clin Oncol 24(1):25–35 43. Bray SJ (2006) Notch signalling: a simple pathway becomes
29. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski complex. Nat Rev Mol Cell Biol 7(9):678–689
RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I, 44. Sainson RC, Harris AL (2008) Regulation of angiogenesis by
Bycott PW, Baum CM, Figlin RA (2007) Sunitinib versus homotypic and heterotypic notch signalling in endothelial cells
interferon alfa in metastatic renal-cell carcinoma. N Engl J Med and pericytes: from basic research to potential therapies.
356(2):115–124 Angiogenesis 11(1):41–51
30. Bollée G, Patey N, Cazajous G, Robert C, Goujon JM, Fakhouri 45. Noguera-Troise I, Daly C, Papadopoulos NJ, Coetzee S, Boland
F, Bruneval P, Noël LH, Knebelmann B (2009) Thrombotic P, Gale NW, Lin HC, Yancopoulos GD, Thurston G (2006)
132 Targ Oncol (2009) 4:121–133

Blockade of Dll4 inhibits tumour growth by promoting non- 63. Nakanishi K, Sweeney W Jr, Avner ED (2001) Segment-specific
productive angiogenesis. Nature 444(7122):1032–1037 c-ErbB2 expression in human autosomal recessive polycystic
46. Pollak MR (2008) Focal segmental glomerulosclerosis: recent kidney disease. J Am Soc Nephrol 12(2):379–384
advances. Curr Opin Nephrol Hypertens 17(2):138–142 64. Mir O, Berveiller P, Ropert S, Goffinet F, Pons G, Treluyer JM,
47. Cheng HT, Kim M, Valerius MT, Surendran K, Schuster-Gossler Goldwasser F (2008) Emerging therapeutic options for breast
K, Gossler A, McMahon AP, Kopan R (2007) Notch2, but not cancer chemotherapy during pregnancy. Ann Oncol 19(4):607–613
Notch1, is required for proximal fate acquisition in the 65. Medina PJ, Goodin S (2008) Lapatinib: a dual inhibitor of
mammalian nephron. Development 134(4):801–811 human epidermal growth factor receptor tyrosine kinases. Clin
48. Niranjan T, Bielesz B, Gruenwald A, Ponda MP, Kopp JB, Ther 30(8):1426–1447
Thomas DB, Susztak K (2008) The Notch pathway in podocytes 66. Cassier PA, Dufresne A, Arifi S, El Sayadi H, Labidi I, Ray-
plays a role in the development of glomerular disease. Nat Med Coquard I, Tabone S, Méeus P, Ranchère D, Sunyach MP,
14(3):290–298 Decouvelaere AV, Alberti L, Blay JY (2008) Imatinib mesilate
49. Kretzler M, Allred L (2008) Notch inhibition reverses kidney for the treatment of gastrointestinal stromal tumour. Expert Opin
failure. Nat Med 14(3):246–247 Pharmacother 9(7):1211–1222
50. Schechter AL, Hung MC, Vaidyanathan L, Weinberg RA, Yang- 67. Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford
Feng TL, Francke U, Ullrich A, Coussens L (1985) The neu JM, Lydon NB, Kantarjian H, Capdeville R, Ohno-Jones S,
gene: an erbB-homologous gene distinct from and unlinked to the Sawyers CL (2001) Efficacy and safety of a specific inhibitor of
gene encoding the EGF receptor. Science 229(4717):976–978 the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N
51. Burgess AW (2008) EGFR family: structure physiology signal- Engl J Med 344(14):1031–1037
ing and therapeutic targets. Growth Factors 26(5):263–274 68. Chevallier P, Hunault-Berger M, Larosa F, Dauriac C, Garand R,
52. Perez EA, Baweja M (2008) HER2-positive breast cancer: Harousseau JL (2008) A phase II trial of high-dose imatinib
current treatment strategies. Cancer Invest 26(6):545–552 mesylate for relapsed or refractory c-kit positive and Bcr-Abl
53. Arany I, Megyesi JK, Kaneto H, Tanaka S, Safirstein RL (2004) negative acute myeloid leukaemia: The AFR-15 trial. Leuk Res .
Activation of ERK or inhibition of JNK ameliorates H(2)O(2) doi:10.1016/j.leukres.2008.09.030
cytotoxicity in mouse renal proximal tubule cells. Kidney Int 69. François H, Coppo P, Hayman JP, Fouqueray B, Mougenot B,
4:1231–1239 Ronco P (2008) Partial fanconi syndrome induced by imatinib
54. Arany I, Megyesi JK, Reusch JE, Safirstein RL (2005) CREB therapy: a novel cause of urinary phosphate loss. Am J Kidney
mediates ERK-induced survival of mouse renal tubular cells after Dis 51(2):298–301
oxidant stress. Kidney Int 68(4):1573–1582 70. Pou M, Saval N, Vera M, Saurina A, Solé M, Cervantes F, Botey
55. Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, A (2003) Acute renal failure secondary to imatinib mesylate
Thongprasert S, Campos D, Maoleekoonpiroj S, Smylie M, Martins treatment in chronic myeloid leukemia. Leuk Lymphoma 44
R, van Kooten M, Dediu M, Findlay B, Tu D, Johnston D, Bezjak (7):1239–1241
A, Clark G, Santabárbara P, Seymour L, National Cancer Institute of 71. Foringer JR, Verani RR, Tjia VM, Finkel KW, Samuels JA,
Canada Clinical Trials Group (2005) Erlotinib in previously treated Guntupalli JS (2005) Acute renal failure secondary to imatinib
non-small cell lung cancer. N Engl J Med 353(2):123–132 mesylate treatment in prostate cancer. Ann Pharmacother 39
56. Miller AA, Murry DJ, Owzar K, Hollis DR, Lewis LD, Kindler (12):2136–2138
HL, Marshall JL, Villalona-Calero MA, Edelman MJ, Hohl RJ, 72. Deininger MW (2008) Nilotinib. Clin Cancer Res 14(13):4027–
Lichtman SM, Ratain MJ (2007) Phase I and pharmacokinetic 4031
study of erlotinib for solid tumors in patients with hepatic or 73. Cortes J, Kim DW, Raffoux E, Martinelli G, Ritchie E, Roy L,
renal dysfunction: CALGB 60101. J Clin Oncol 25(21):3055– Coutre S, Corm S, Hamerschlak N, Tang JL, Hochhaus A,
3060 Khoury HJ, Brümmendorf TH, Michallet M, Rege-Cambrin G,
57. Masutani K, Fujisaki K, Maeda H, Toyonaga J, Inoshima I, Gambacorti-Passerini C, Radich JP, Ernst T, Zhu C, Van Tornout
Takayama K, Katafuchi R, Hirakata H, Tsuruya K, Iida M (2008) JM, Talpaz M (2008) Efficacy and safety of dasatinib in
Tubulointerstitial nephritis and IgA nephropathy in a patient with imatinib-resistant or -intolerant patients with chronic myeloid
advanced lung cancer treated with long-term gefitinib. Clin Exp leukemia in blast phase. Leukemia 22(12):2176–2183
Nephrol 12(5):398–402 74. Comoglio PM, Giordano S, Trusolino L (2008) Drug develop-
58. Kumasaka R, Nakamura N, Shirato K, Osawa H, Takanashi S, ment of MET inhibitors: targeting oncogene addiction and
Hasegawa Y, Yamabe H, Nakamura M, Tamura M, Okumura K expedience. Nat Rev Drug Discov 7(6):504–516
(2004) Side effects of therapy: case 1. Nephrotic syndrome 75. Liu Y, Yang J (2006) Hepatocyte growth factor: new arsenal in
associated with gefitinib therapy. J Clin Oncol 22(12):2504–2505 the fights against renal fibrosis? Kidney Int 70(2):238–240
59. Lautrette A, Li S, Alili R, Sunnarborg SW, Burtin M, Lee DC, 76. Srinivasan R, Choueiri TK, Vaishampayan U, Rosenberg JE,
Friedlander G, Terzi F (2005) Angiotensin II and EGF receptor Stein MN, Logan T, Bukowski RM, Mueller T, Keer HN,
cross-talk in chronic kidney diseases: a new therapeutic Linehan WM (2008) A phase II study of the dual MET/VEGFR2
approach. Nat Med 11(8):867–874 inhibitor XL880 in patients (pts) with papillary renal carcinoma
60. Patel DK (2008) Clinical use of anti-epidermal growth factor (PRC). J Clin Oncol 26(May 20 suppl):5103 abstract
receptor monoclonal antibodies in metastatic colorectal cancer. 77. Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D
Pharmacotherapy 28(11 Pt 2):31S–41S (1999) A more accurate method to estimate glomerular filtration
61. Giro C, Berger B, Bölke E, Ciernik IF, Duprez F, Locati L, rate from serum creatinine: a new prediction equation. Modifi-
Maillard S, Ozsahin M, Pfeffer R, Robertson AG, Langendijk cation of Diet in Renal Disease Study Group. Ann Intern Med
JA, Budach W (2009) High rate of severe radiation dermatitis 130(6):461–470
during radiation therapy with concurrent cetuximab in head and 78. Launay-Vacher V, Oudard S, Janus N, Gligorov J, Pourrat X,
neck cancer: results of a survey in EORTC institutes. Radiother Rixe O, Morere JF, Beuzeboc P, Deray G (2007) Renal
Oncol 90(2):166–171 insufficiency and cancer medications (IRMA) study group.
62. Nielsen DL, Andersson M, Kamby C (2009) HER2-targeted Prevalence of renal insufficiency in cancer patients and implica-
therapy in breast cancer. Monoclonal antibodies and tyrosine tions for anticancer drug management: the renal insufficiency and
kinase inhibitors. Cancer Treat Rev 35(2):121–136 anticancer medications (IRMA) study. Cancer 110(6):1376–1384
Targ Oncol (2009) 4:121–133 133

79. Kintzel PE (2001) Anticancer drug-induced kidney disorders. Small EJ (2008) Bevacizumab plus interferon alfa compared
Drug Saf 24(1):19–38 with interferon alfa monotherapy in patients with metastatic renal
80. Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE, cell carcinoma: CALGB 90206. J Clin Oncol 26(33):5422–5428
Campbell BT, Chan KW, Ciceri P, Davis MI, Edeen PT, Faraoni 92. Bukowski RM (2008) Randomized trial of bevacizumab in
R, Floyd M, Hunt JP, Lockhart DJ, Milanov ZV, Morrison MJ, advanced renal cell carcinoma. Curr Oncol Rep 10(2):99–100
Pallares G, Patel HK, Pritchard S, Wodicka LM, Zarrinkar PP 93. Johnson DH, Fehrenbacher L, Novotny WF, Herbst RS,
(2008) A quantitative analysis of kinase inhibitor selectivity. Nat Nemunaitis JJ, Jablons DM, Langer CJ, DeVore RF 3rd,
Biotechnol 26(1):127–132 Gaudreault J, Damico LA, Holmgren E, Kabbinavar F (2004)
81. Izzedine H, Brocheriou I, Deray G, Rixe O (2007) Thrombotic Randomized phase II trial comparing bevacizumab plus carbo-
microangiopathy and anti-VEGF agents. Nephrol Dial Trans- platin and paclitaxel with carboplatin and paclitaxel alone in
plant 22(5):1481–1482 previously untreated locally advanced or metastatic non-small-
82. Scartozzi M, Galizia E, Chiorrini S, Giampieri R, Berardi R, cell lung cancer. J Clin Oncol 22(11):2184–2191
Ierantoni C, Cascinu S (2009) Arterial hypertension correlates 94. Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati
with clinical outcome in colorectal cancer patients treated with A, Lilenbaum R, Johnson DH (2006) Paclitaxel-carboplatin
first-line bevacizumab. Ann Oncol 20(2):227–230 alone or with bevacizumab for non-small-cell lung cancer. N
83. Rixe O, Billemont B, Izzedine H (2007) Hypertension as a Engl J Med 355(24):2542–2550
predictive factor of Sunitinib activity. Ann Oncol 18(6):1117 95. Herbst RS, Johnson DH, Mininberg E, Carbone DP, Henderson
84. Rini BI, Schiller JH, Fruehauf JP, Cohen EE, Tarazi JC, T, Kim ES, Blumenschein G Jr, Lee JJ, Liu DD, Truong MT,
Rosbrook B, Ricart AD, Olszanski AJ, Kim S, Spano J (2008) Hong WK, Tran H, Tsao A, Xie D, Ramies DA, Mass R,
Association of diastolic blood pressure (dBP) ≥90 mmHg with Seshagiri S, Eberhard DA, Kelley SK, Sandler A (2005) Phase I/
overall survival (OS) in patients treated with axitinib (AG- II trial evaluating the anti-vascular endothelial growth factor
013736). J Clin Oncol 26(May 20 suppl):3543 abstract monoclonal antibody bevacizumab in combination with the
85. Miller KD, Chap LI, Holmes FA, Cobleigh MA, Marcom PK, HER-1/epidermal growth factor receptor tyrosine kinase inhib-
Fehrenbacher L, Dickler M, Overmoyer BA, Reimann JD, Sing itor erlotinib for patients with recurrent non-small-cell lung
AP, Langmuir V, Rugo HS (2005) Randomized phase III trial of cancer. J Clin Oncol 23(11):2544–2555
capecitabine compared with bevacizumab plus capecitabine in 96. Herbst RS, O’Neill VJ, Fehrenbacher L, Belani CP, Bonomi PD,
patients with previously treated metastatic breast cancer. J Clin Hart L, Melnyk O, Ramies D, Lin M, Sandler A (2007) Phase II
Oncol 23(4):792–799 study of efficacy and safety of bevacizumab in combination with
86. Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, chemotherapy or erlotinib compared with chemotherapy alone
Shenkier T, Cella D, Davidson NE (2007) Paclitaxel plus for treatment of recurrent or refractory non small-cell lung
bevacizumab versus paclitaxel alone for metastatic breast cancer. cancer. J Clin Oncol 25(30):4743–4750
N Engl J Med 357(26):2666–2676 97. Hochster HS, Hart LL, Ramanathan RK, Childs BH, Hainsworth
87. Dickler MN, Rugo HS, Eberle CA, Brogi E, Caravelli JF, Panageas JD, Cohn AL, Wong L, Fehrenbacher L, Abubakr Y, Saif MW,
KS, Boyd J, Yeh B, Lake DE, Dang CT, Gilewski TA, Bromberg JF, Schwartzberg L, Hedrick E (2008) Safety and efficacy of
Seidman AD, D’Andrea GM, Moasser MM, Melisko M, Park JW, oxaliplatin and fluoropyrimidine regimens with or without
Dancey J, Norton L, Hudis CA (2008) A phase II trial of erlotinib in bevacizumab as first-line treatment of metastatic colorectal
combination with bevacizumab in patients with metastatic breast cancer: results of the TREE study. J Clin Oncol 26(21):3523–
cancer. Clin Cancer Res 14(23):7878–7883 3529
88. Dellapasqua S, Bertolini F, Bagnardi V, Campagnoli E, Scarano 98. Saltz LB, Clarke S, Diaz-Rubio E, Scheithauer W, Figer A,
E, Torrisi R, Shaked Y, Mancuso P, Goldhirsch A, Rocca A, Wong R, Koski S, Lichinitser M, Yang TS, Rivera F, Couture F,
Pietri E, Colleoni M (2008) Metronomic cyclophosphamide and Sirzen F, Cassidy J (2008) Bevacizumab in combination with
capecitabine combined with bevacizumab in advanced breast oxaliplatin-based chemotherapy as first-line therapy in metastatic
cancer. J Clin Oncol 26(30):4899–4905 colorectal cancer: a randomized phase III study. J Clin Oncol 26
89. Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, (12):2013–2019
Topalian SL, Steinberg SM, Chen HX, Rosenberg SA (2003) A 99. Tol J, Koopman M, Rodenburg CJ, Cats A, Creemers GJ,
randomized trial of bevacizumab, an anti-vascular endothelial Schrama JG, Erdkamp FL, Vos AH, Mol L, Antonini NF, Punt
growth factor antibody, for metastatic renal cancer. N Engl J Med CJ (2008) A randomised phase III study on capecitabine,
349(5):427–434 oxaliplatin and bevacizumab with or without cetuximab in first-
90. Escudier B, Pluzanska A, Koralewski P, Ravaud A, Bracarda S, line advanced colorectal cancer, the CAIRO2 study of the dutch
Szczylik C, Chevreau C, Filipek M, Melichar B, Bajetta E, colorectal cancer group (DCCG). an interim analysis of toxicity.
Gorbunova V, Bay JO, Bodrogi I, Jagiello-Gruszfeld A, Moore Ann Oncol 19(4):734–738
N, AVOREN Trial investigators (2007) Bevacizumab plus 100. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T,
interferon alfa-2a for treatment of metastatic renal cell carcino- Hainsworth J, Heim W, Berlin J, Baron A, Griffing S,
ma: A randomised, double-blind phase III trial. Lancet 370 Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R,
(9605):2103–2111 Kabbinavar F (2004) Bevacizumab plus irinotecan, fluoroura-
91. Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Ou cil, and leucovorin for metastatic colorectal cancer. N Engl J
SS, Archer L, Atkins JN, Picus J, Czaykowski P, Dutcher J, Med 350(23):2335–2342

You might also like