Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/7430852

Renal-Cell Carcinoma

Article  in  New England Journal of Medicine · January 2006


DOI: 10.1056/NEJMra043172 · Source: PubMed

CITATIONS READS
1,349 2,341

2 authors, including:

Herbert T Cohen
Boston University
56 PUBLICATIONS   3,608 CITATIONS   

SEE PROFILE

All content following this page was uploaded by Herbert T Cohen on 28 May 2014.

The user has requested enhancement of the downloaded file.


The new england journal of medicine

review article

medical progress

Renal-Cell Carcinoma
Herbert T. Cohen, M.D., and Francis J. McGovern, M.D.

i n the united states, renal cancer is the 7th leading malignant


condition among men and the 12th among women, accounting for 2.6 percent of
all cancers.1 About 2 percent of cases of renal cancer are associated with inherited
syndromes. In the United States, 36,160 new cases of renal cancer are predicted to oc-
cur in 2005, many of which are being discovered earlier because of the widespread
From the Renal and Hematology–Oncolo-
gy Sections, Departments of Medicine and
Pathology, Boston University School of
Medicine (H.T.C.); and the Department of
Urology, Massachusetts General Hospital
and Harvard Medical School (F.J.M.) —
all in Boston. Address reprint requests to
availability of radiographic testing. Nevertheless, 12,660 deaths from the disease are Dr. Cohen at the Department of Medicine,
predicted to occur in 2005.1 Renal-cell carcinomas arise from the renal epithelium and Renal Section, Boston University School of
account for about 85 percent of renal cancers. A quarter of the patients present with ad- Medicine, Evans Biomedical Research Cen-
ter, 650 Albany St., Rm. X-535, Boston, MA
vanced disease, including locally invasive or metastatic renal-cell carcinoma. More- 02118, or at htcohen@bu.edu.
over, a third of the patients who undergo resection of localized disease will have a re-
currence. Median survival for patients with metastatic disease is about 13 months. N Engl J Med 2005;353:2477-90.
Copyright © 2005 Massachusetts Medical Society.
Thus, there is a great need for more effective surgical and medical therapies.

overview
The classic presentation of renal-cell carcinoma includes the triad of flank pain, hema-
turia, and a palpable abdominal mass. Few patients now present in this manner.
Roughly half the cases are now detected because a renal mass is incidentally identified
on radiographic examination. Other common presenting features may be nonspecific,
such as fatigue, weight loss, or anemia. Risk factors for renal-cell carcinoma include
smoking, obesity, and hypertension,2 as well as acquired cystic kidney disease associ-
ated with end-stage renal disease. A 1.6:1.0 male predominance exists,1 and the peak
incidence is in the sixth and seventh decades. Gross or microscopic hematuria is an im-
portant clinical clue to the diagnosis of renal-cell carcinoma; thus, hematuria should
be evaluated promptly by a computed tomographic (CT) scan of the genitourinary tract
and, in patients older than 40 years of age, by cystoscopy to rule out bladder cancer.
Prognosis is closely related to the stage of disease (Fig. 1). The Heidelberg classifica-
tion of renal tumors was introduced in 19976 as a means of more completely correlat-
ing the histopathological features with the identified genetic defects (Table 1).

tumor types and molecular pathogenesis


conventional or clear-cell renal-cell carcinoma
Von Hippel–Lindau disease (number 193300 in Mendelian Inheritance in Man [MIM])
is a rare, autosomal dominant, familial cancer syndrome consisting chiefly of retinal
angiomas, hemangioblastomas of the central nervous system, pheochromocytomas,
and renal-cell carcinoma of the clear-cell type (Fig. 2). The von Hippel–Lindau tumor-
suppressor gene (VHL) was identified in 1993.7 In this disease, one VHL allele is inher-
ited with a mutation. Associated focal lesions, such as renal-cell carcinoma, arise from
the inactivation or silencing of the remaining normal (wild-type) VHL allele (Fig. 3). Re-
markably, defects in the VHL gene also appear to be responsible for about 60 percent of

n engl j med 353;23 www.nejm.org december 8, 2005 2477

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

Figure 1. Staging Overview and Five-Year Survival Rates for Renal Cancer.
Survival data3 are based on the 1997 tumor–node–metastasis (TNM) staging guidelines.4 More recent renal-cancer
staging is described elsewhere.5

the cases of sporadic clear-cell renal-cell carcino- that promotes the ubiquitination and destruction
ma,8 which represents a major portion of all cases of proteins (Fig. 4B).12,13 VHL protein binds the
of renal-cell carcinoma. transcriptional activators hypoxia-inducible factor
VHL protein, the product of the VHL gene, func- 1a (HIF-1a) and 2a (HIF-2a) directly and destabiliz-
tions as a tumor suppressor, inhibiting growth es them.14 Furthermore, VHL protein promotes the
when reintroduced into cultures of renal-cell carci- ubiquitination and destruction of HIF-a.15-17 These
noma.9,10 Hypoxia-inducible genes are normally in- VHL-regulated pathways are being studied as po-
hibited by VHL protein,11 including several encod- tential targets of therapies for clear-cell renal-cell
ing proteins involved in angiogenesis (e.g., vascular carcinoma.
endothelial growth factor [VEGF]), cell growth HIF is the key regulator of the hypoxic response
(e.g., transforming growth factor a [TGF-a]), glu- in multicellular organisms. Thus, VHL protein has
cose uptake (e.g., the GLUT-1 glucose transporter), a central role in oxygen sensing. For HIF-a to bind
and acid–base balance (e.g., carbonic anhydrase IX VHL protein, a proline residue must undergo hy-
[CA9]). When VHL protein is lost, these proteins are droxylation, which is an unusual protein modifi-
overexpressed, creating a microenvironment favor- cation18,19 (Fig. 4B). A family of proline hydroxy-
able for epithelial-cell proliferation (Fig. 4A). Thus, lases operates on HIF-a in a graded fashion, so that
cells deficient in VHL protein behave as if they are hy- the extent of hydroxylation depends on oxygen ten-
poxic, even in conditions of normoxia. VHL protein, sion.20,21 Hydroxylation of an asparagine residue
with elongin proteins C and B, binds cul2 protein (a blocks the interaction of HIF-a with the transcrip-
member of the cullin family of ubiquitin ligase pro- tional coactivator p300.22 Thus, multiple hydroxyl-
teins), indicating that some VHL protein serves as ation steps cooperate to inhibit HIF-a activity.
the receptor subunit of a ubiquitin ligase complex To correlate the genotype with the disease phe-

2478 n engl j med 353;23 www.nejm.org december 8 , 2005

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

Table 1. Sporadic and Hereditary Renal-Cell Carcinomas and Genetic Defects According to Histologic Appearance.*

Sporadic Renal-Cell Carcinomas Renal-Cell Carcinomas in an Inherited Syndrome


Histologic Appearance Incidence Gene and Frequency Rare Syndrome† Gene
percent
Conventional 75 VHL, 60 VHL disease VHL
FCRC Chromosome 3p translocation
Hereditary paraganglioma SDHB
Papillary 12 MET, 13 HPRC MET
TFE3, <1 HLRCC FH
Chromophobe 4 Birt–Hogg–Dubé syndrome BHD
Oncocytoma 4 Birt–Hogg–Dubé syndrome BHD
Collecting duct <1
Unclassified 3–5

* VHL denotes von Hippel–Lindau, FCRC familial clear-cell renal cancer, SDHB succinate dehydrogenase B, HPRC hered-
itary papillary renal carcinoma, HLRCC hereditary leiomyomatosis and renal-cell cancer, and FH fumarate hydratase.
† Additional rare syndromes or infrequent associations are not included.

notype, naturally occurring VHL mutations have ing 5:1 male predominance. Localized papillary re-
been evaluated to determine their effect on HIF-a nal-cell carcinoma metastasizes less frequently than
ubiquitination. An intriguing finding is that the clear-cell renal-cell carcinoma.34 However, the sur-
VHL mutations that disrupt HIF-a processing are vival rate for metastatic papillary renal-cell carci-
the same as those associated with the vascular man- noma is probably worse than that for clear-cell
ifestations of von Hippel–Lindau disease, such as renal-cell carcinoma.35 The risk of both types is
hemangioblastoma (Fig. 2).15,16,23,24 Since renal- particularly increased among patients with end-
cell carcinoma develops in only a subgroup of pa- stage renal disease. Chromosome 7, which harbors
tients with hemangioblastoma, the overexpression the MET proto-oncogene, is duplicated in 75 percent
of HIF-a appears to be necessary for, but not suf- of sporadic papillary cases. There are two subtypes
ficient to induce, renal tumorigenesis. Neverthe- of papillary renal-cell carcinoma.36 Type 1 tumors
less, HIF-a is vitally important to the pathogenesis are papillary lesions covered by small cells with pale
of this disease. VHL-induced inhibition of HIF-a cytoplasm and small oval nuclei with indistinct
is sufficient to suppress the growth of clear-cell nucleoli, and type 2 tumors are papillary lesions
renal-cell carcinoma in preclinical models.25,26 The covered by large cells with abundant eosinophilic
cell-matrix protein fibronectin,27 chaperonin TRiC/ cytoplasm. Type 2 cells are typified by pseudostratifi-
CCT,28 microtubules,29 and transcription factor cation and large, spherical nuclei with distinct nu-
Jade-130-32 are all molecules that interact with VHL cleoli. Type 2 tumors are genetically more heteroge-
protein in a manner that is dependent on VHL mu- neous, have a poorer prognosis, and may arise from
tation, suggesting that they may also contribute to type 1 tumors.37
disease pathogenesis. Papillary renal-cell carcinoma occurs in several
Distinct from von Hippel–Lindau disease, famil- familial syndromes (MIM number 605074). He-
ial clear-cell renal cancer has been reported in pa- reditary papillary renal carcinoma is an autosomal
tients with translocations of chromosome 3p at a dominant disorder associated with multifocal pap-
fragile site at 3p14.33 Loss of the translocated chro- illary renal-cell carcinoma38 with type 1 histologic
mosome 3p probably implicates VHL protein in features (Fig. 3).39 The causative gene, mutations in
the development of these tumors. Additional trans- which are responsible for hereditary papillary re-
locations of chromosome 3 have been associated nal carcinoma, has been identified at chromo-
with clear-cell renal-cell carcinoma as well. some 7 and encodes MET, a receptor tyrosine kinase
that is normally activated by hepatocyte growth
papillary renal-cell carcinoma factor 40 (Fig. 4C). In hereditary papillary renal car-
Sporadic papillary renal-cell carcinoma has a five- cinoma, the MET receptor tyrosine kinase domain
year survival rate approaching 90 percent and a strik- undergoes autoactivating amino-acid–substitu-

n engl j med 353;23 www.nejm.org december 8, 2005 2479

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

Figure 2. Schematic Representation of the Clinical Spectrum of von Hippel–Lindau Disease and Potential Biologic Mechanisms.
The three major manifestations of von Hippel–Lindau (VHL) disease — central nervous system hemangioblastoma, pheochromocytoma,
and clear-cell renal-cell carcinoma — are represented as Venn diagrams. Affected families may not have all three conditions, depending large-
ly on the type of mutation in the VHL gene inherited. Circle sizes are not intended to reflect the numbers of patients in each group. Type 1 VHL
disease (Panel A) is most common; it comprises clear-cell renal-cell carcinoma with central nervous system hemangioblastoma and is due to
major disruptions in the VHL protein, such as those resulting from truncating mutations. Type 2 VHL disease (Panel A) includes pheochro-
mocytoma with one, both, or neither of the other features. Type 2B disease predisposes patients to all three conditions, whereas type 2A in-
cludes hemangioblastoma and pheochromocytoma and type 2C includes only pheochromocytoma. Type 2 is most often due to more subtle
VHL gene mutations, such as missense mutations that result in the substitution of a single amino acid. Besides classic von Hippel–Lindau
disease, sporadic cases of hemangioblastoma or pheochromocytoma have been shown to be due to germ-line VHL mutations. In addition,
familial Chuvash polycythemia, which does not include any of the features of von Hippel–Lindau disease, is an autosomal recessive disorder
caused by homozygosity for the specific VHL missense mutation R200W. In Panel B, the spectrum of manifestations of von Hippel–Lindau
disease, along with biochemical studies, suggests the involvement of specific biochemical pathways. For example, the mutations in VHL pro-
tein that disrupt the ubiquitination and destruction of hypoxia-inducible factor a (HIF-a) are the same as those that correlate with the lesions
of hemangioblastoma. Interestingly, von Hippel–Lindau disease type 2A mutations disrupt HIF-a processing and do not cause renal cancer,
suggesting that the HIF pathway plus additional, unknown VHL pathways (called X pathways) must be disrupted for clear-cell renal-cell car-
cinoma to develop. The probable involvement of additional pathways is supported by the multistep nature of renal tumorigenesis. Alterna-
tively, the unknown pathway or pathways may simply reflect a higher level of expression of HIF-a in this subgroup of patients. Pheochromocy-
tomas can arise from mutations in the gene for VHL protein that do not affect HIF-a processing, suggesting that they arise through the
disruption of other, unknown VHL pathways (called Y pathways). Thus, the overexpression of HIF-a correlates closely with the development
of hemangioblastoma; appears to be necessary for, but not sufficient to induce, renal tumorigenesis; and is not necessary for the develop-
ment of pheochromocytoma.

tion mutations, which promote cellular transforma- type 2 histologic features.44 Occasionally, cases of
tion.41 Subsequently, chromosome 7 harboring the collecting-duct or clear-cell renal-cell carcinoma
MET mutation is duplicated, increasing the gene occur. These cases of papillary renal-cell carcino-
dose.42,43 Only a small percentage of the cases of ma metastasize early and are the most aggressive
the sporadic papillary type have MET mutations.40 of the familial types.45 Intriguingly, FH, the gene that
Thus, the pathogenesis of hereditary papillary re- causes this autosomal dominant syndrome, encodes
nal carcinoma is usually different from that of spo- fumarate hydratase, a Krebs-cycle enzyme.46 As with
radic papillary renal-cell carcinoma. the loss of a tumor-suppressor gene, the wild-type
Patients with the hereditary leiomyomatosis and FH allele is lost in hereditary leiomyomatosis and
renal-cell cancer syndrome (MIM number 605839) in lesions of renal-cell carcinoma.47 Along simi-
are at risk for cutaneous and uterine leiomyo- lar lines, cases of renal-cell carcinoma with solid
mas and solitary papillary renal-cell carcinoma with histologic features or cases of the clear-cell form

2480 n engl j med 353;23 www.nejm.org december 8 , 2005

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

Figure 3. Steps in the Development of Renal-Cell Carcinoma.


In contrast to sporadic renal-cell carcinoma (Panels A and C), fewer steps are required for the development of renal-cell carcinoma in the in-
herited forms of the disease (Panels B and D), because all of the patient’s cells have a mutation that predisposes the patient to the disease.
As a result, the disease associated with the familial syndromes occurs earlier and is often multifocal. Each familial renal cancer syndrome is
autosomal dominant. In von Hippel–Lindau disease, a cellular recessive mechanism is involved, since both copies of the VHL gene are inac-
tivated (Panels A and B). VHL is a classic tumor-suppressor gene. In hereditary papillary renal carcinoma, one copy of the MET gene has an
activating mutation, which is inherited (Panel D). Chromosome 7, which includes the defective MET allele, becomes duplicated, increasing
the level of expression of the activated MET protein, which is a receptor tyrosine kinase for hepatocyte growth factor. Activated MET is a clas-
sic oncogene. A plus sign represents the wild-type allele; a minus sign represents a null allele. A plus sign in red type represents a mutated,
activated allele; two plus signs in red type represent duplication of that allele.

n engl j med 353;23 www.nejm.org december 8, 2005 2481

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

have been reported in patients with the hereditary


paraganglioma syndrome (MIM number 115310). Figure 4 (facing page). Molecular Mechanisms of the
Development of Renal-Cell Carcinoma.
Certain forms of hereditary paraganglioma are as-
Pathologic cooperativity between renal-cancer cells
sociated with germ-line defects in the succinate de- of the clear-cell type and adjacent vasculature is shown
hydrogenase B gene.48 Succinate dehydrogenase B in Panel A. In clear-cell renal-cell carcinoma, hypoxia-
protein is another mitochondrial, Krebs-cycle en- inducible factor a (HIF-a) transcription factor accumu-
zyme. Thus, an intriguing connection exists among lates, resulting in the overexpression of proteins that are
cellular ATP production, the hypoxic response, and normally inducible with hypoxia, such as transforming
growth factor a and b (TGF-a and TGF-b, respectively),
tumorigenesis in both neuronal and kidney tissue. vascular endothelial growth factor (VEGF), and platelet-
A number of sporadic cases of papillary renal- derived growth factor B chain (PDGF-B). The overex-
cell carcinoma have chromosomal translocations in- pressed VEGF, PDGF-B, and TGF-b act on neighboring
volving the TFE3 gene at chromosome Xp11.2.49-51 vascular cells to promote tumor angiogenesis. The aug-
Children and young adults are affected without mented tumor vasculature provides additional nutrients
and oxygen to promote the growth of tumor cells. TGF-a
predilection for sex, and the histologic features of acts in an autocrine manner on the tumor cells by signal-
such cases have been variably described as papil- ing through the epidermal growth factor receptor, which
lary renal-cell carcinoma, clear-cell renal-cell carci- promotes tumor-cell proliferation and survival. The role
noma, or a unique type of pathology. The TFE3 gene of von Hippel–Lindau (VHL) protein in clear-cell renal-
encodes a helix–loop–helix transcription factor cell carcinoma and in controlling the expression of the
HIF-a transcription factors is shown in Panel B. Under
related to the proto-oncogene product c-myc. Key normoxic conditions HIF-a is hydroxylated on two pro-
TRE3 domains become fused with other gene prod- line residues by a proline hydroxylase and on an aspar-
ucts, and renal-cell carcinoma is probably due to agine residue by an asparagine hydroxylase.
TFE3 overexpression. Hydroxylation (OH) by proline hydroxylase permits bind-
ing of HIF-a to VHL protein, which promotes the ubiquit-
ination (Ub) and destruction of HIF-a by the proteasome
oncocytoma and chromophobe renal-cell pathway. Hydroxylation by asparagine hydroxylase blocks
carcinoma the interaction of HIF-a with transcriptional coactivator
Oncocytomas, which are benign, account for about p300. VHL protein, with elongin proteins C and B, binds
4 percent of nephrectomies performed because re- cul2 protein (a member of the cullin family of ubiquitin
nal-cell carcinoma is suspected. The chromophobe ligase proteins). RING-box protein Rbx1 serves as the
ubiquitin transferase for the VHL skp-cullin-F-box pro-
variant of renal-cell carcinoma also accounts for tein (SCF) complex. In the absence of wild-type VHL
4 percent of all cases of renal-cell carcinoma52 and protein, hydroxylated HIF-a accumulates and is able
may have a benign course after surgery, provided to heterodimerize with HIF-b and activate transcription
that the tumor stage and grade are favorable.53 On- at hypoxia-response elements (HREs), which are found
cocytoma is thought to originate from type A in- in genes such as VEGF. In hypoxic conditions, HIF-a is
not hydroxylated and so cannot bind VHL protein. Panel
tercalated cells of the collecting duct, whereas C shows the role of MET in papillary renal carcinoma.
chromophobe renal-cell carcinoma is thought to MET is a receptor tyrosine kinase for hepatocyte growth
originate from type B intercalated cells. factor. In the absence of ligand, MET normally exists in
The Birt–Hogg–Dubé syndrome (MIM number an autoinhibited state. MET homodimerizes in the pres-
135150) is a rare autosomal dominant disorder ence of ligand hepatocyte growth factor and undergoes
reciprocal phosphorylation and activation. In hereditary
characterized by hair-follicle hamartomas (fibro- papillary renal carcinoma and in occasional sporadic
folliculomas) of the face and neck.54-57 About 15 papillary renal-cell carcinoma, activating mutations of
percent of affected patients have multiple renal MET disinhibit the receptor, even in the absence of
tumors, most often chromophobe or mixed chro- ligand. Furthermore, the mutated MET allele on chromo-
mophobe–oncocytomas. Occasionally, papillary or some 7 becomes duplicated, increasing the expression
of the activated MET protein.
clear-cell renal-cell carcinoma develops in patients
with the Birt–Hogg–Dubé syndrome. BHD, the gene
implicated in the syndrome, encodes the protein fol-
liculin,58 a suspected tumor suppressor. BHD mu- collecting-duct renal-cell carcinoma
tations occur only rarely in sporadic renal-cell car- Collecting-duct renal-cell carcinoma accounts for
cinoma.59,60 The Birt–Hogg–Dubé renal phenotype less than 1 percent of all cases of renal-cell carcino-
supports the existence of a close relationship be- ma and is typically an aggressive tumor. Medullary
tween oncocytoma and chromophobe renal-cell carcinoma of the kidney, which may be a variant of
carcinoma. the collecting-duct type, is associated with sickle

2482 n engl j med 353;23 www.nejm.org december 8 , 2005

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

n engl j med 353;23 www.nejm.org december 8, 2005 2483

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

cell trait or disease. The collecting-duct form may ing. Organizations such as the VHL Family Alli-
be most similar to transitional-cell carcinoma of the ance (www.vhl.org) are a vital resource for patients,
urothelium. families, physicians, and researchers.

management of sporadic prognosis


and hereditary renal-cell
carcinoma Defining the prognosis of renal-cell carcinoma is
important for both therapeutic decision-making
An enhancing renal mass on a CT scan obtained and counseling patients. For metastatic renal-cell
after the administration of contrast material is a carcinoma, poor prognostic factors include a low
strong clue that renal cancer is present. A staging Karnofsky performance-status score (a standard
workup should be performed before treatment is way of measuring functional impairment in pa-
initiated. Multiple enhancing lesions, or a family his- tients with cancer), a high level of serum lactate de-
tory of renal-cell carcinoma, particularly in persons hydrogenase, a low hemoglobin level, and a high
younger than 50 years of age, suggests a hereditary corrected level of serum calcium.63,64 The Univer-
predisposition to the disease. Von Hippel–Lindau sity of California, Los Angeles, Integrated Staging
disease, hereditary leiomyomatosis and renal-cell System was developed to evaluate the prognosis at
cancer, and the Birt–Hogg–Dubé syndrome all have diagnosis and in the presence of metastatic dis-
extrarenal manifestations, whereas familial clear- ease; it includes tumor–node–metastasis (TNM)
cell renal cancer and hereditary papillary renal carci- staging, the patient’s score on the Eastern Cooper-
noma do not. Thus, a careful physical examination ative Oncology Group performance-status scale (an-
including ophthalmologic, neurologic, and derma- other measure of functional impairment in patients
tologic evaluation may be helpful. CT scanning or with cancer), and the Fuhrman nuclear grade, which
magnetic resonance imaging (MRI) of the abdomen assesses histologic features of the tumor.65 This sys-
and pelvis may reveal uterine tumors in patients tem has been used successfully in more than 4000
with hereditary leiomyomatosis and renal-cell can- patients at eight international centers.66
cer or renal cysts or pancreatic or adrenal involve-
ment in patients with von Hippel–Lindau disease. surgical treatment
Patients with hereditary renal-cell carcinoma
should be closely monitored. CT before and after radical nephrectomy
the administration of contrast material is the best Surgical excision is the primary treatment for re-
test for detection and assessment of renal masses, nal-cell carcinoma. Radical nephrectomy, which
with gadolinium-enhanced MRI as an alternative. includes removal of the kidney en bloc with Gero-
Such studies can be performed at intervals ranging ta’s fascia, the ipsilateral adrenal gland, and re-
from every three to six months to every two to three gional lymph nodes, has been the standard thera-
years, depending on the size of the lesions and the py, although more limited approaches are being
type of syndrome. Larger masses require more fre- explored. The surgical approach is determined by
quent evaluation. Because small masses are usually the size and location of the tumor within the kid-
of low grade, they can be observed until they reach ney, the TNM stage, and any special anatomical
3 cm, at which time they should be removed.61,62 considerations.
However, tumors caused by hereditary leiomyoma- Staging and evaluation for the presence of me-
tosis and renal-cell cancer should be excised im- tastases, including a careful history-taking and
mediately because of their aggressive nature. Pa- physical examination, should be completed before
tients with von Hippel–Lindau disease should surgery. Routine laboratory studies should include
undergo MRI studies of the brain and spinal cord measurement of the hematocrit and serum levels
to screen for hemangioblastoma. A family pedigree of creatinine, calcium, and alkaline phosphatase and
should be generated, and family members at risk a urinalysis for proteinuria. Imaging studies, such as
should be encouraged to seek medical attention. radiographs of the chest, CT of the abdomen and
Testing is available for the VHL, MET, FH, and BHD pelvis, and in some cases, MRI evaluation of the re-
genes. One goal of such testing is to free unaffect- nal vein and inferior vena cava, CT of the chest or
ed family members from continued cancer screen- head, or bone scanning may be needed. The fre-

2484 n engl j med 353;23 www.nejm.org december 8, 2005

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

quency of follow-up after surgery depends on the adjacent structures. Because identification of the
stage of the tumor. type of renal-cell carcinoma is important, a core bi-
opsy of the renal mass should be performed as part
surgery for metastatic disease of the procedure. Ideal candidates for minimally
Nephrectomy may be warranted, even in the pres- invasive percutaneous ablative therapy are patients
ence of metastatic disease. The combination of in- with tumors less than 3 cm in diameter who have
terferon alfa and nephrectomy is superior to inter- serious coexisting conditions and for whom stan-
feron alfa alone, offering a survival advantage of dard approaches would pose substantial risks. Pa-
3 to 10 months.67,68 Surgical excision of a solitary tients with multifocal tumors may also benefit from
metastasis in patients with advanced renal-cell car- minimally invasive percutaneous procedures. High-
cinoma is recommended in many cases, but this frequency focused ultrasound applied externally to
approach has not yet been proved to be effective in the body is being studied as another potential min-
prolonging survival. imally invasive therapy.

nephron-sparing partial nephrectomy medical treatment


Nephron-sparing partial nephrectomy has gained
acceptance for treating tumors less than 4 cm in di- Medical therapies are generally offered for locally
ameter. Other indications for partial nephrectomy advanced or metastatic renal-cell carcinoma (Ta-
may include a solitary kidney, bilateral renal masses, ble 2), and much of the clinical experience with
or renal insufficiency, as well as the presence of hy- this approach is in patients with the clear-cell type.
pertension, diabetes, or hereditary renal-cell carci- Because response rates are low, the need to identify
noma syndromes. Results achieved with nephron- new therapeutic agents is great.74
sparing surgery are similar to those with radical
nephrectomy, but a disadvantage is a rate of local chemotherapy
recurrence of 3 to 6 percent.69 Rates of response to chemotherapy alone are low
(roughly 4 to 6 percent).75 Drug resistance may be
laparoscopic nephrectomy related to the expression of the multidrug resis-
First reported in 1991,70 laparoscopic nephrecto- tance transporter in proximal-tubule cells — the
my has accelerated the evolution toward minimally cells from which clear-cell and papillary renal-cell
invasive surgical management of renal-cell carci- carcinoma may originate. Chemotherapy may be
noma. The benefits of the laparoscopic approach more efficacious for advanced non–clear-cell re-
include decreased postoperative pain, a shorter hos- nal-cell carcinoma, particularly the collecting-duct
pitalization, and a quicker recovery. The laparo- type.76-78 A phase 2 trial of carboplatin and pacli-
scopic approach has been used for both radical taxel for the collecting-duct form of the disease is
nephrectomy and partial nephrectomy. The laparo- under way.
scopic partial nephrectomy, however, is a technically
demanding procedure with the potential for in- immunomodulatory therapies
creased perioperative complications.71 The value of immunomodulatory therapy for clear-
cell renal-cell carcinoma is supported by reports of
percutaneous ablative approaches occasional spontaneous tumor regression, infre-
The most recent evolution in the surgical manage- quent complete regression of metastatic disease
ment of small tumors has been percutaneous ther- with cytokine therapies, and promising early results
mal ablative techniques that use radiofrequency with allogeneic stem-cell transplantation and tumor
heat ablation72 or cryoablation73 to destroy tumor vaccines. The goal of immunomodulatory therapy
cells. A needle probe is advanced through the skin is to boost either tumor antigenicity or host surveil-
and directed into the tumor under image guid- lance. Unique tumor antigens may also be induc-
ance. Although early results of radiofrequency ab- ible in renal-cell carcinoma.79
lation and cryoablation are encouraging, larger tri-
als with long-term follow-up are needed. The rates Interferon Alfa
of complications appear to be low, but reported ad- About 14 percent of cases of metastatic clear-cell
verse events include intraoperative and postopera- renal carcinoma respond to interferon alfa alone.
tive hemorrhage, urinary leakage, and injury to Various doses and routes have been used.80 The me-

n engl j med 353;23 www.nejm.org december 8, 2005 2485

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

of patients with metastatic disease. Identifying fea-


Table 2. Medical Therapies for Advanced Renal Cancer.*
tures predictive of a response to interleukin-2 would
FDA-approved regimen represent a further advance, and efforts are being
High-dose interleukin-2 (aldesleukin, immunomodula- made to identify patients with clear-cell renal carci-
tory cytokine)
noma who would be likely to have a response to in-
Commonly used agents terleukin-2 therapy on the basis of pathological
Low-dose interleukin-2
characteristics and expression of CA9.
Interferon alfa (immunomodulatory cytokine)
Experimental therapies adjuvant therapy
Bevacizumab (humanized VEGF-neutralizing antibody)
Given the high rate of recurrence of renal-cell car-
Sunitinib malate (VEGF receptor and multitargeted kinase
inhibitor) cinoma after nephrectomy, a follow-up adjuvant
Sorafenib tosylate (VEGF receptor and multitargeted ki- approach would be desirable, especially for pa-
nase inhibitor) tients with high-risk, locally advanced disease.
Panitumumab (human EGFR-neutralizing antibody) However, conventional chemotherapy, interferon
Gefitinib (EGFR tyrosine kinase inhibitor) alfa, or even interleukin-283 has not proved effec-
Erlotinib (EGFR tyrosine kinase inhibitor)
tive as an adjuvant therapy. Approaches currently
Temsirolimus (inhibitor of the mammalian target of
rapamycin) being tested include tumor vaccines and a mono-
Tumor vaccines clonal antibody directed against CA9.
Allogeneic stem-cell transplantation
evolving therapies
* FDA denotes Food and Drug Administration, VEGF vas-
cular endothelial growth factor, and EGFR epidermal stem-cell transplantation
growth factor receptor. The trade name for interleukin-2
is Proleukin; for bevacizumab, Avastin; for sunitinib Allogeneic stem-cell transplantation performed af-
malate, Sutent; for sorafenib tosylate, Nexavar; for gefi- ter the administration of a non–marrow ablative
tinib, Iressa; for erlotinib, Tarceva. regimen elicits a potent graft-versus-tumor effect
and appears promising for treating clear-cell renal-
cell carcinoma.84 Protocols developed at the Nation-
dian duration of response is six months and rarely al Institutes of Health have used myelosuppress-
exceeds two years. Because the side effects of the ive pretreatment, followed by an infusion of donor
drug are not onerous, it appears to be a good choice CD34+ cells and T cells from an HLA-identical sib-
to use in combination with other agents in experi- ling.84 A course of immunosuppressive agents,
mental approaches. such as cyclosporine, is used to limit graft-versus-
host disease and is rapidly tapered. Twenty of the
Interleukin-2 first 45 patients with metastatic renal-cell carcino-
High-dose interleukin-2 is the standard therapy for ma who underwent stem-cell transplantation had a
advanced renal-cell carcinoma and is the only regi- response (44 percent).85 However, results in some
men for this disease approved by the Food and other centers have been less promising. The re-
Drug Administration. However, many patients with sponses have correlated well with the development
metastatic disease cannot take high-dose interleu- of graft-versus-host disease and with the conver-
kin-2, because it causes a capillary leak syndrome sion of T-cell chimerism to full donor origin. One
or because it is not available in all treatment cen- goal is to identify the tumor epitopes that are initi-
ters. High-dose interleukin-2 induces responses in ating the graft-versus-tumor response to improve
21 percent of patients, as compared with only 13 treatment specificity. The two drawbacks to stem-
percent of patients who receive low-dose interleu- cell transplantation have been severe graft-versus-
kin-2.81 The median duration of response has been host disease, which can be life-threatening, and
reported to be 54 months overall, and for those the need for a haplotype-matched sibling donor.
with a complete response, the median duration of a Prognosis is also an important guide to patient se-
response is yet to be reached.82 Interleukin-2 has lection, since responses take several months. The
also been used in combination with other drugs, next generation of strategies for stem-cell transplan-
but it is unclear whether combined therapy achieves tation may include the use of tumor vaccines after
better results than interleukin-2 alone. Thus, inter- transplantation as well as the use of cytokine thera-
leukin-2 is a highly effective therapy for a subgroup py to boost recipient or even donor immunity.

2486 n engl j med 353;23 www.nejm.org december 8, 2005

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

tumor vaccines vided a key “proof of principle” of the efficacy of


Tumor vaccines represent a potential means of en- anti-angiogenic therapy and may offer additional
hancing host immunity. A promising approach to benefit when given in combination with other
the treatment of advanced clear-cell renal carcinoma drugs. Inhibitors of VEGF receptor tyrosine kinase
uses autologous or donor dendritic cells, which are being developed and tested. Indeed, the multi-
initiate a primary immune response by presenting targeted kinase inhibitors sunitinib and sorafenib
antigen in the context of costimulatory molecules. have shown great promise in phase 2 and phase 3
Dendritic cells can be pulsed with tumor protein,86 trials, with at least stabilization of disease in as many
DNA, or RNA87; they can even be fused with tumor as 70 percent of patients with cytokine-refractory
cells88,89 to present tumor antigens in a context fa- disease.
vorable for therapy. Such vaccines are generally
well tolerated, but they will require further optimi- TGF-a–Pathway Components as Molecular Targets
zation. Concomitant administration of cytokines TGF-a is a potent growth factor for epithelial cells
may improve the response to vaccines. that acts through the epidermal growth factor re-
ceptor (EGFR), which is a receptor tyrosine kinase.
target antigens TGF-a is overexpressed in the epithelium in clear-
A goal of stem-cell or vaccine therapies is to char- cell renal carcinoma and is a VHL target.95 Over-
acterize the tumor antigens involved in the immune expression of TGF-a is an early event in the patho-
response. One potential target is the G250 renal can- genesis of this disease.96 Furthermore, growth
cer antigen, which has been identified as CA9. The of renal cancer cells in culture is dependent on
CA9 gene is a target of HIF and so is overexpressed TGF-a.97 Thus, the TGF-a pathway is a logical
in VHL-related clear-cell renal carcinoma, even in the choice for therapeutic intervention.
earliest lesions of von Hippel–Lindau disease.90 Antibodies against EGFR are thought to bind
Thus, in cases of renal-cell carcinoma, a high pro- EGFR and promote its down-regulation from
portion of CA9-positive cells may be associated with the cell surface. A fully human monoclonal anti-
a more favorable prognosis.91 As a transmembrane body against human EGFR, called panitumumab
protein, CA9 may also be a therapeutically useful tu- (ABX-EGF), has been evaluated in a phase 2 trial
mor antigen.92,93 It will be important to identify involving 88 patients with metastatic renal-cell car-
additional target antigens. cinoma.98 Only one patient had a complete re-
sponse, and two had partial responses — a disap-
therapies targeting vegf and tgf-a pointing result. Small-molecule inhibitors of the
pathways EGFR tyrosine kinase are also being developed.99
Originally identified as regulated by VHL, VEGF The quinazolines gefitinib and erlotinib are now in
and TGF-a are now promising therapeutic targets phase 2 trials. In a phase 1 trial of erlotinib, just
in clear-cell renal carcinoma. The manner in which one patient with metastatic disease had a com-
these molecules interact with the cancer epitheli- plete response.100 Erlotinib is also being tested in
um and surrounding vascular endothelium leads to combination with bevacizumab, although encour-
tumor progression (Fig. 4A). A combination of ther- aging initial results could not be confirmed in a
apies based on rational targets such as these may randomized phase 2 trial.
therefore be a powerful approach to advanced renal-
cell carcinoma. Other Approaches
Temsirolimus (CCI-779), a selective inhibitor of
VEGF-Pathway Components as Molecular Targets the mammalian target of rapamycin, has shown
VEGF is overexpressed throughout clear-cell renal- efficacy in a phase 2 trial of metastatic renal-cell
cell carcinoma tissue and may be the most impor- carcinoma.101 Temsirolimus may inhibit HIF as
tant tumor angiogenic factor. A randomized phase well. Partial responses were noted in 7 percent of
2 trial involving patients with metastatic renal-cell patients, and minor responses in 26 percent. The
carcinoma investigated the efficacy of bevacizu- median survival rate was 15 months. The notable
mab, a humanized VEGF-neutralizing antibody.94 activity of the drug in patients with poor prognos-
This agent extended the interval before tumor pro- tic features prompted a phase 3 trial. Other op-
gression to 4.8 months, as compared with 2.5 tions are being pursued, including agents target-
months for placebo. Bevacizumab therefore pro- ing HIF.

n engl j med 353;23 www.nejm.org december 8, 2005 2487

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

The mutated, activated hepatocyte growth factor


summary and prospects
for the future receptor MET could be targeted in the papillary
form of the disease.102,103 The immune respon-
The poor prognosis of advanced renal-cell carci- siveness of renal-cell carcinoma provides an op-
noma demands an aggressive search for new ther- portunity for the development and optimization of
apeutic agents and strategies. Leads will probably vaccines and other immune therapies. Preserva-
come from both a careful elucidation of the biology tion of as much renal function as possible and re-
of each type of the disease and broader approach- duced rates of complications are two goals of new
es, such as gene-expression–array and proteome minimally invasive approaches to renal-cell carci-
analyses. Much has been accomplished since the noma; other goals are to identify early markers of
identification of the VHL gene in 1993. Already, disease, prognosis, or responsiveness to therapy.
VHL protein pathways, such as those involving Supported in part by grants (RO1 CA79830 and RO1 DK67569, to
VEGF and TGF-a, are being exploited therapeuti- Dr. Cohen) from the National Institutes of Health.
We are indebted to Drs. Michael B. Atkins, William G. Kaelin, Jr.,
cally, and agents affecting these pathways might be Matthew R. Smith, Walter M. Stadler, and Berton Zbar for careful re-
more effective when used in combination. Identi- view of the manuscript.
fication of the MET gene was another key advance.

refer enc es
1. Jemal A, Murray T, Ward E, et al. Cancer proteins. Proc Natl Acad Sci U S A 1997;94: 22. Lando D, Peet DJ, Whelan DA, Gorman
statistics, 2005. CA Cancer J Clin 2005;55: 2156-61. JJ, Whitelaw ML. Asparagine hydroxylation
10-30. 13. Lonergan KM, Iliopoulos O, Ohh M, et of the HIF transactivation domain a hypoxic
2. Chow WH, Gridley G, Fraumeni JF Jr, al. Regulation of hypoxia-inducible mRNAs switch. Science 2002;295:858-61.
Jarvholm B. Obesity, hypertension, and the by the von Hippel-Lindau tumor suppressor 23. Clifford SC, Cockman ME, Smallwood
risk of kidney cancer in men. N Engl J Med protein requires binding to complexes con- AC, et al. Contrasting effects on HIF-1alpha
2000;343:1305-11. taining elongins B/C and Cul2. Mol Cell Biol regulation by disease-causing pVHL muta-
3. Javidan J, Stricker HJ, Tamboli P, et al. 1998;18:732-41. tions correlate with patterns of tumourigen-
Prognostic significance of the 1997 TNM 14. Maxwell PH, Wiesener MS, Chang GW, esis in von Hippel-Lindau disease. Hum Mol
classification of renal cell carcinoma. J Urol et al. The tumour suppressor protein VHL Genet 2001;10:1029-38.
1999;162:1277-81. targets hypoxia-inducible factors for oxy- 24. Hoffman MA, Ohh M, Yang H, Klco JM,
4. Kidney. In: Fleming ID, Cooper JS, Hen- gen-dependent proteolysis. Nature 1999; Ivan M, Kaelin WG Jr. von Hippel-Lindau
son DE, et al., eds. AJCC cancer staging hand- 399:271-5. protein mutants linked to type 2C VHL dis-
book. Philadelphia: Lippincott-Raven, 1998: 15. Cockman ME, Masson N, Mole DR, et ease preserve the ability to downregulate
215-7. al. Hypoxia inducible factor-alpha binding HIF. Hum Mol Genet 2001;10:1019-27.
5. Kidney. In: Greene FL, Page DL, Fleming and ubiquitylation by the von Hippel-Lindau 25. Kondo K, Kim WY, Lechpammer M,
ID, et al., eds. AJCC cancer staging manual. tumor suppressor protein. J Biol Chem 2000; Kaelin WG Jr. Inhibition of HIF2alpha is
New York: Springer-Verlag, 2002:323-5. 275:25733-41. sufficient to suppress pVHL-defective tumor
6. Kovacs G, Akhtar M, Beckwith BJ, et al. 16. Ohh M, Park CW, Ivan M, et al. Ubiquit- growth. PLoS Biol 2003;1:E83.
The Heidelberg classification of renal cell ination of hypoxia-inducible factor requires 26. Zimmer M, Doucette D, Siddiqui N, Il-
tumours. J Pathol 1997;183:131-3. direct binding to the beta-domain of the von iopoulos O. Inhibition of hypoxia-inducible
7. Latif F, Tory K, Gnarra J, et al. Identifica- Hippel-Lindau protein. Nat Cell Biol 2000; factor is sufficient for growth suppression
tion of the von Hippel-Lindau disease tumor 2:423-7. of VHL¡/¡ tumors. Mol Cancer Res 2004;2:
suppressor gene. Science 1993;260:1317-20. 17. Tanimoto K, Makino Y, Pereira T, Poel- 89-95.
8. Kim WY, Kaelin WG. Role of VHL gene linger L. Mechanism of regulation of the 27. Ohh M, Yauch RL, Lonergan KM, et al.
mutation in human cancer. J Clin Oncol hypoxia-inducible factor-1 alpha by the von The von Hippel-Lindau tumor suppressor
2004;22:4991-5004. Hippel-Lindau tumor suppressor protein. protein is required for proper assembly of
9. Iliopoulos O, Kibel A, Gray S, Kaelin EMBO J 2000;19:4298-309. an extracellular fibronectin matrix. Mol Cell
WG Jr. Tumour suppression by the human 18. Ivan M, Kondo K, Yang H, et al. HIF- 1998;1:959-68.
von Hippel-Lindau gene product. Nat Med alpha targeted for VHL-mediated destruc- 28. Feldman DE, Spiess C, Howard DE,
1995;1:822-6. tion by proline hydroxylation: implications Frydman J. Tumorigenic mutations in VHL
10. Chen F, Kishida T, Duh FM, et al. Sup- for O2 sensing. Science 2001;292:464-8. disrupt folding in vivo by interfering with
pression of growth of renal carcinoma cells 19. Jaakkola P, Mole DR, Tian YM, et al. Tar- chaperonin binding. Mol Cell 2003;12:
by the von Hippel-Lindau tumor suppressor geting of HIF-alpha to the von Hippel-Lindau 1213-24.
gene. Cancer Res 1995;55:4804-7. ubiquitylation complex by O2-regulated pro- 29. Hergovich A, Lisztwan J, Barry R,
11. Iliopoulos O, Levy AP, Jiang C, Kaelin lyl hydroxylation. Science 2001;292:468-72. Ballschmieter P, Krek W. Regulation of mi-
WG Jr, Goldberg MA. Negative regulation of 20. Epstein AC, Gleadle JM, McNeill LA, et crotubule stability by the von Hippel-Lindau
hypoxia-inducible genes by the von Hippel- al. C. elegans EGL-9 and mammalian ho- tumour suppressor protein pVHL. Nat Cell
Lindau protein. Proc Natl Acad Sci U S A mologs define a family of dioxygenases that Biol 2003;5:64-70.
1996;93:10595-9. regulate HIF by prolyl hydroxylation. Cell 30. Zhou MI, Wang H, Ross JJ, Kuzmin I, Xu
12. Pause A, Lee S, Worrell RA, et al. The 2001;107:43-54. C, Cohen HT. The von Hippel-Lindau tumor
von Hippel-Lindau tumor-suppressor gene 21. Bruick RK, McKnight SL. A conserved suppressor stabilizes novel plant homeo-
product forms a stable complex with human family of prolyl-4-hydroxylases that modify domain protein Jade-1. J Biol Chem 2002;
CUL-2, a member of the Cdc53 family of HIF. Science 2001;294:1337-40. 277:39887-98.

2488 n engl j med 353;23 www.nejm.org december 8, 2005

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

31. Zhou MI, Wang H, Foy RL, Ross JJ, Co- al. Germline mutations in FH predispose to 61. Walther MM, Choyke PL, Glenn G, et al.
hen HT. Tumor suppressor von Hippel- dominantly inherited uterine fibroids, skin Renal cancer in families with hereditary re-
Lindau (VHL) stabilization of Jade-1 protein leiomyomata and papillary renal cell cancer. nal cancer: prospective analysis of a tumor
occurs through plant homeodomains and is Nat Genet 2002;30:406-10. size threshold for renal parenchymal spar-
VHL mutation dependent. Cancer Res 2004; 47. Kiuru M, Launonen V, Hietala M, et al. ing surgery. J Urol 1999;161:1475-9.
64:1278-86. Familial cutaneous leiomyomatosis is a two- 62. Herring JC, Enquist EG, Chernoff A,
32. Zhou MI, Foy RL, Chitalia VC, et al. hit condition associated with renal cell can- Linehan WM, Choyke PL, Walther MM. Pa-
Jade-1, a candidate renal tumor suppressor cer of characteristic histopathology. Am J renchymal sparing surgery in patients with
that promotes apoptosis. Proc Natl Acad Sci Pathol 2001;159:825-9. hereditary renal cell carcinoma: 10-year ex-
U S A 2005;102:11035-40. 48. Vanharanta S, Buchta M, McWhinney perience. J Urol 2001;165:777-81.
33. Cohen AJ, Li FP, Berg S, et al. Hereditary SR, et al. Early-onset renal cell carcinoma as 63. Motzer RJ, Mazumdar M, Bacik J, Berg W,
renal-cell carcinoma associated with a chro- a novel extraparaganglial component of Amsterdam A, Ferrara J. Survival and prog-
mosomal translocation. N Engl J Med 1979; SDHB-associated heritable paraganglioma. nostic stratification of 670 patients with ad-
301:592-5. Am J Hum Genet 2004;74:153-9. vanced renal cell carcinoma. J Clin Oncol
34. Beck SD, Patel MI, Snyder ME, et al. Ef- 49. Meloni AM, Dobbs RM, Pontes JE, Sand- 1999;17:2530-40.
fect of papillary and chromophobe cell type berg AA. Translocation (X;1) in papillary re- 64. Motzer RJ, Bacik J, Schwartz LH, et al.
on disease-free survival after nephrectomy nal cell carcinoma: a new cytogenetic sub- Prognostic factors for survival in previously
for renal cell carcinoma. Ann Surg Oncol type. Cancer Genet Cytogenet 1993;65:1-6. treated patients with metastatic renal cell
2004;11:71-7. 50. Sidhar SK, Clark J, Gill S, et al. The carcinoma. J Clin Oncol 2004;22:454-63.
35. Motzer RJ, Bacik J, Mariani T, Russo P, t(X;1)(p11.2;q21.2) translocation in papil- 65. Zisman A, Pantuck AJ, Dorey F, et al. Im-
Mazumdar M, Reuter V. Treatment outcome lary renal cell carcinoma fuses a novel gene proved prognostication of renal cell carcino-
and survival associated with metastatic re- PRCC to the TFE3 transcription factor gene. ma using an integrated staging system.
nal cell carcinoma of non-clear-cell histolo- Hum Mol Genet 1996;5:1333-8. J Clin Oncol 2001;19:1649-57.
gy. J Clin Oncol 2002;20:2376-81. 51. Weterman MA, Wilbrink M, Geurts van 66. Patard JJ, Kim HL, Lam JS, et al. Use of
36. Delahunt B, Eble JN. Papillary renal cell Kessel A. Fusion of the transcription fac- the University of California Los Angeles in-
carcinoma: a clinicopathologic and immu- tor TFE3 gene to a novel gene, PRCC, in tegrated staging system to predict survival in
nohistochemical study of 105 tumors. Mod t(X;1)(p11;q21)-positive papillary renal cell renal cell carcinoma: an international multi-
Pathol 1997;10:537-44. carcinomas. Proc Natl Acad Sci U S A 1996; center study. J Clin Oncol 2004;22:3316-22.
37. Gunawan B, von Heydebreck A, Fritsch 93:15294-8. 67. Flanigan RC, Salmon SE, Blumenstein
T, et al. Cytogenetic and morphologic typing 52. Cheville JC, Lohse CM, Zincke H, Weav- BA, et al. Nephrectomy followed by interfer-
of 58 papillary renal cell carcinomas: evidence er AL, Blute ML. Comparisons of outcome on alfa-2b compared with interferon alfa-2b
for a cytogenetic evolution of type 2 from and prognostic features among histologic alone for metastatic renal-cell cancer. N Engl
type 1 tumors. Cancer Res 2003;63:6200-5. subtypes of renal cell carcinoma. Am J Surg J Med 2001;345:1655-9.
38. Zbar B, Glenn G, Lubensky I, et al. He- Pathol 2003;27:612-24. 68. Mickisch GH, Garin A, van Poppel H,
reditary papillary renal cell carcinoma: clini- 53. Peyromaure M, Misrai V, Thiounn N, de Prijck L, Sylvester R. Radical nephrecto-
cal studies in 10 families. J Urol 1995;153: et al. Chromophobe renal cell carcinoma: my plus interferon-alfa-based immunother-
907-12. analysis of 61 cases. Cancer 2004;100:1406- apy compared with interferon alfa alone in
39. Lubensky IA, Schmidt L, Zhuang Z, et al. 10. metastatic renal-cell carcinoma: a random-
Hereditary and sporadic papillary renal car- 54. Birt AR, Hogg GR, Dube WJ. Hereditary ised trial. Lancet 2001;358:966-70.
cinomas with c-met mutations share a dis- multiple fibrofolliculomas with trichodisco- 69. Novick AC. Nephron-sparing surgery
tinct morphological phenotype. Am J Pathol mas and acrochordons. Arch Dermatol 1977; for renal cell carcinoma. Annu Rev Med 2002;
1999;155:517-26. 113:1674-7. 53:393-407.
40. Schmidt L, Duh FM, Chen F, et al. 55. Weirich G, Glenn G, Junker K, et al. Fa- 70. Clayman RV, Kavoussi LR, Soper NJ, et
Germline and somatic mutations in the ty- milial renal oncocytoma: clinicopathologi- al. Laparoscopic nephrectomy: initial case
rosine kinase domain of the MET proto- cal study of 5 families. J Urol 1998;160:335- report. J Urol 1991;146:278-82.
oncogene in papillary renal carcinomas. Nat 40. 71. Gill IS, Matin SF, Desai MM, et al. Com-
Genet 1997;16:68-73. 56. Toro JR, Glenn G, Duray P, et al. Birt- parative analysis of laparoscopic versus open
41. Jeffers M, Schmidt L, Nakaigawa N, et Hogg-Dube syndrome: a novel marker of partial nephrectomy for renal tumors in 200
al. Activating mutations for the met tyrosine kidney neoplasia. Arch Dermatol 1999;135: patients. J Urol 2003;170:64-8.
kinase receptor in human cancer. Proc Natl 1195-202. 72. Zlotta AR, Wildschutz T, Raviv G, et al.
Acad Sci U S A 1997;94:11445-50. 57. Zbar B, Alvord WG, Glenn G, et al. Risk Radiofrequency interstitial tumor ablation
42. Zhuang Z, Park WS, Pack S, et al. Tri- of renal and colonic neoplasms and sponta- (RITA) is a possible new modality for treat-
somy 7-harbouring non-random duplica- neous pneumothorax in the Birt-Hogg- ment of renal cancer: ex vivo and in vivo ex-
tion of the mutant MET allele in hereditary Dube syndrome. Cancer Epidemiol Bio- perience. J Endourol 1997;11:251-8.
papillary renal carcinomas. Nat Genet 1998; markers Prev 2002;11:393-400. 73. Uchida M, Imaide Y, Sugimoto K, Ueha-
20:66-9. 58. Nickerson ML, Warren MB, Toro JR, et ra H, Watanabe H. Percutaneous cryosur-
43. Fischer J, Palmedo G, von Knobloch R, al. Mutations in a novel gene lead to kidney gery for renal tumours. Br J Urol 1995;75:
et al. Duplication and overexpression of the tumors, lung wall defects, and benign tumors 132-6.
mutant allele of the MET proto-oncogene in of the hair follicle in patients with the Birt- 74. Motzer RJ. Renal cell carcinoma: a pri-
multiple hereditary papillary renal cell tu- Hogg-Dube syndrome. Cancer Cell 2002;2: ority malignancy for development and study
mours. Oncogene 1998;17:733-9. 157-64. of novel therapies. J Clin Oncol 2003;21:
44. Launonen V, Vierimaa O, Kiuru M, et al. 59. Khoo SK, Kahnoski K, Sugimura J, et al. 1193-4.
Inherited susceptibility to uterine leiomyo- Inactivation of BHD in sporadic renal tu- 75. Yagoda A, Abi-Rached B, Petrylak D.
mas and renal cell cancer. Proc Natl Acad Sci mors. Cancer Res 2003;63:4583-7. Chemotherapy for advanced renal-cell car-
U S A 2001;98:3387-92. 60. da Silva NF, Gentle D, Hesson LB, Mor- cinoma: 1983-1993. Semin Oncol 1995;22:
45. Zbar B, Klausner R, Linehan WM. Study- ton DG, Latif F, Maher ER. Analysis of the 42-60.
ing cancer families to identify kidney cancer Birt-Hogg-Dube (BHD) tumour suppressor 76. Gollob JA, Upton MP, DeWolf WC, At-
genes. Annu Rev Med 2003;54:217-33. gene in sporadic renal cell carcinoma and co- kins MB. Long-term remission in a patient
46. Tomlinson IP, Alam NA, Rowan AJ, et lorectal cancer. J Med Genet 2003;40:820-4. with metastatic collecting duct carcinoma

n engl j med 353;23 www.nejm.org december 8, 2005 2489

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

treated with taxol/carboplatin and surgery. cinoma with tumor lysate-pulsed autolo- tumor suppressor. Cancer Res 1998;58:226-
Urology 2001;58:1058. gous dendritic cells. Clin Cancer Res 2002; 31.
77. Peyromaure M, Thiounn N, Scotte F, 8:3369-76. 96. Everitt JI, Walker CL, Goldsworthy TW,
Vieillefond A, Debre B, Oudard S. Collecting 87. Su Z, Dannull J, Heiser A, et al. Immu- Wolf DC. Altered expression of transform-
duct carcinoma of the kidney: a clinicopath- nological and clinical responses in meta- ing growth factor-alpha: an early event in re-
ological study of 9 cases. J Urol 2003;170: static renal cancer patients vaccinated with nal cell carcinoma development. Mol Car-
1138-40. tumor RNA-transfected dendritic cells. Can- cinog 1997;19:213-9.
78. Milowsky MI, Rosmarin A, Tickoo SK, cer Res 2003;63:2127-33. 97. de Paulsen N, Brychzy A, Fournier MC,
Papanicolaou N, Nanus DM. Active chemo- 88. Marten A, Renoth S, Heinicke T, et al. et al. Role of transforming growth factor-al-
therapy for collecting duct carcinoma of the Allogeneic dendritic cells fused with tumor pha in von Hippel–Lindau (VHL)(¡/¡) clear
kidney: a case report and review of the litera- cells: preclinical results and outcome of a cell renal carcinoma cell proliferation: a pos-
ture. Cancer 2002;94:111-6. clinical phase I/II trial in patients with meta- sible mechanism coupling VHL tumor sup-
79. Hanada K, Yewdell JW, Yang JC. Im- static renal cell carcinoma. Hum Gene Ther pressor inactivation and tumorigenesis.
mune recognition of a human renal cancer 2003;14:483-94. Proc Natl Acad Sci U S A 2001;98:1387-92.
antigen through post-translational protein 89. Avigan D, Vasir B, Gong J, et al. Fusion 98. Rowinsky EK, Schwartz GH, Gollob JA,
splicing. Nature 2004;427:252-6. cell vaccination of patients with metastatic et al. Safety, pharmacokinetics, and activity
80. Small EJ, Motzer RJ. Interferon for renal breast and renal cancer induces immuno- of ABX-EGF, a fully human anti-epidermal
cell carcinoma. In: Belldegrun A, Ritchie logical and clinical responses. Clin Cancer growth factor receptor monoclonal antibody
AWS, Figlin RA, Oliver RTD, Vaughan ED, Res 2004;10:4699-708. in patients with metastatic renal cell cancer.
eds. Renal and adrenal tumors. New York: 90. Mandriota SJ, Turner KJ, Davies DR, et J Clin Oncol 2004;22:3003-15.
Oxford University Press, 2003:381-7. al. HIF activation identifies early lesions in 99. Arteaga CL. Overview of epidermal
81. Yang JC, Sherry RM, Steinberg SM, et al. VHL kidneys: evidence for site-specific tumor growth factor receptor biology and its role
Randomized study of high-dose and low- suppressor function in the nephron. Cancer as a therapeutic target in human neoplasia.
dose interleukin-2 in patients with meta- Cell 2002;1:459-68. Semin Oncol 2002;29:Suppl:3-9.
static renal cancer. J Clin Oncol 2003;21: 91. Bui MH, Seligson D, Han KR, et al. Car- 100. Hidalgo M, Siu LL, Nemunaitis J, et al.
3127-32. bonic anhydrase IX is an independent pre- Phase I and pharmacologic study of OSI-
82. Fisher RI, Rosenberg SA, Fyfe G. Long- dictor of survival in advanced renal clear cell 774, an epidermal growth factor receptor ty-
term survival update for high-dose recombi- carcinoma: implications for prognosis and rosine kinase inhibitor, in patients with ad-
nant interleukin-2 in patients with renal cell therapy. Clin Cancer Res 2003;9:802-11. vanced solid malignancies. J Clin Oncol 2001;
carcinoma. Cancer J Sci Am 2000;6:Suppl 1: 92. Luiten RM, Coney LR, Fleuren GJ, War- 19:3267-79.
S55-S57. naar SO, Litvinov SV. Generation of chimeric 101. Atkins MB, Hidalgo M, Stadler WM, et
83. Clark JI, Atkins MB, Urba WJ, et al. Ad- bispecific G250/anti-CD3 monoclonal anti- al. Randomized phase II study of multiple
juvant high-dose bolus interleukin-2 for pa- body, a tool to combat renal cell carcinoma. dose levels of CCI-779, a novel mammalian
tients with high-risk renal cell carcinoma: Br J Cancer 1996;74:735-44. target of rapamycin kinase inhibitor, in pa-
a cytokine working group randomized trial. 93. Steffens MG, Boerman OC, de Mulder tients with advanced refractory renal cell car-
J Clin Oncol 2003;21:3133-40. PH, et al. Phase I radioimmunotherapy of cinoma. J Clin Oncol 2004;22:909-18.
84. Childs R, Chernoff A, Contentin N, et al. metastatic renal cell carcinoma with 131I- 102. Lynch TJ, Bell DW, Sordella R, et al. Ac-
Regression of metastatic renal-cell carcino- labeled chimeric monoclonal antibody G250. tivating mutations in the epidermal growth
ma after nonmyeloablative allogeneic periph- Clin Cancer Res 1999;5:3268s-3274s. factor receptor underlying responsiveness
eral-blood stem-cell transplantation. N Engl 94. Yang JC, Haworth L, Sherry RM, et al. of non–small-cell lung cancer to gefitinib.
J Med 2000;343:750-8. A randomized trial of bevacizumab, an anti- N Engl J Med 2004;350:2129-39.
85. Srinivasan R, Barrett J, Childs R. Alloge- vascular endothelial growth factor antibody, 103. Paez JG, Janne PA, Lee JC, et al. EGFR
neic stem cell transplantation as immuno- for metastatic renal cancer. N Engl J Med mutations in lung cancer: correlation with
therapy for nonhematological cancers. Sem- 2003;349:427-34. clinical response to gefitinib therapy. Sci-
in Oncol 2004;31:47-55. 95. Knebelmann B, Ananth S, Cohen HT, ence 2004;304:1497-500.
86. Holtl L, Zelle-Rieser C, Gander H, et al. Sukhatme VP. Transforming growth factor Copyright © 2005 Massachusetts Medical Society.
Immunotherapy of metastatic renal cell car- alpha is a target for the von Hippel-Lindau

apply for jobs electronically at the nejm careercenter


Physicians registered at the NEJM CareerCenter can apply for jobs electronically using
their own cover letters and CVs. You can keep track of your job-application history with
a personal account that is created when you register with the CareerCenter and apply
for jobs seen online at our Web site. Visit www.nejmjobs.org for more information.

2490 n engl j med 353;23 www.nejm.org december 8, 2005

Downloaded from www.nejm.org by ADITI SINGH MD on January 14, 2009 .


Copyright © 2005 Massachusetts Medical Society. All rights reserved.
View publication stats

You might also like