Urologic Clinics of North America
Urologic Clinics of North America
Urologic Clinics of North America
Preface
in causing remission of metastatic disease and regression of primary tumors with prolonged survival
achieved in patients formerly facing a grave prognosis. In this issue of the Urologic Clinics of North
America numerous experts in this field present
a contemporary understanding of the epidemiology, pathology, molecular biology, radiology, and
clinical management of renal cortical tumors with
an encouraging eye toward the future.
A historical view of kidney surgery reveals the
dangers and high mortality rates associated with
total nephrectomy in the latter years of the nineteenth century (>40%) and the beginning of the
twentieth century (>20%) until the time of the urologist Edwin Beer in 1920 (<4%). In the first half of
the twentieth century, patients were only diagnosed with large, symptomatic (palpable masses
with hematuria and pain), and usually locally advanced renal tumors, which provided a surgical
challenge to the urologists. At that time, incidentally detected renal tumors represented less than
5% of the total. Charles Robsons published experience with transthoracic radical nephrectomy (including ipsilateral adrenal, all perinephric soft
tissues, and regional retroperitoneal lymph nodes)
in 1963 provided the mainstay surgical approach
to all renal tumors during the pre-CT era until
the mid 1980s when elective kidney-sparing
operations were first described. A perioperative
mortality rate of 3% was further improved on as
postWorld War II methods of anesthesia, blood
banking, and perioperative care were integrated
into the surgical management of renal tumors.
The discovery and widespread use of the modern abdominal imaging techniques (CT, MRI, and
abdominal ultrasound) over the last 2 decades
changed the profile of the renal tumor patient
medwedi.ru
urologic.theclinics.com
Guest Editor
xiv
Preface
from one who had a massive, symptomatic tumor
at presentation to the current patient who has
a small, asymptomatic renal mass (<4 cm) incidentally discovered in 70% of the cases after
evaluation of nonspecific abdominal and musculoskeletal pain or during unrelated cancer diagnosis
and follow-up care. At the same time as this clinical size and stage migration was occurring, an enhanced understanding of the pathology of renal
tumors was underway. Until the 1980s, renal cortical tumors were classified only by descriptive
terms, such as clear, granular, papillary, sarcomatoid, and renal oncocytoma. The realization that
the renal cortical tumors were, in fact, members
of a complex family with unique pathologic appearances, cytogenetic and molecular defects, familial and hereditary syndromes, and variable
metastatic potentials lead to the development of
the current Heidelberg Classification of renal tumors in 1997. We now know that granular and sarcomatoid tumors are not specific histologic
subtypes, whereas chromophobe, papillary, conventional clear cell, medullary, collecting duct,
metanephric adenoma, and oncocytoma are.
Approximately 90% of the renal tumors that are
metastatic have the conventional clear cell
histology.
Focused research into hereditary and familial tumor syndromes, which account for less than 5% of
all renal cortical tumors, also provided molecular
insight into the sporadic cases within each of the
histologic subtypes. Investigators from the National Cancer Institute discovered that the tumor
suppressor gene VHL, located at chromosome
3p25, could be inactivated by various mutations,
loss of heterozygosity, hypermethylations, or alterations in VHL modifier genes, and was responsible
for the von Hippel-Lindau syndrome. This autosomal dominant syndrome is characterized by retinal
hemangiomas, conventional clear cell renal cell
carcinoma, renal cysts, cerebellar and spinal
hemangioblastomas, pheochromocytomas, endocrine pancreatic tumors, and epididymal cysts.
The dysfunction of the VHL protein increases hypoxia-induced factor (HIF) 1 alpha resulting in
a marked overexpression of vascular and endothelial growth factors and the hypervascular state
observed in the VHL syndrome of tumors in general and its associated conventional clear cell carcinomas of the kidney. Loss of VHL gene function
also occurs in at least 50% of sporadic conventional clear cell carcinomas. Similar intensive
research into other hereditary renal tumor syndromes, including hereditary papillary RCC (cMet
mutation at chromosome 7q34), hereditary leiomyomatosis syndrome and renal cell carcinoma
(HLRCC, mutation in fumarate hydratase gene),
and Birt Hogge Dube syndrome of fibrofolliculomas, chromophobe and oncocytic predominant
renal tumors, pneumothorax, and bronchiectasis
(recently mapped to 17q12) has provided tremendous insight into the molecular mechanisms by
which these tumors operate. Similar cytogenetic
and molecular defects were observed in the histologic subtypes of the sporadic forms of renal
cancer. The research and development of new
systemic agents, tyrosine kinase and mTOR inhibitors, which specifically target pathways that stimulate renal tumor regulation and growth, allowed
for clinical trials in metastatic renal cell carcinoma
with marked improvement in response rates
compared with traditional chemotherapy and
cytokines. In addition, tumor-specific immunohistochemical stains and molecular probes can
now precisely diagnose and subclassify renal tumors far better than traditional pathologic
assessment.
Modern imaging with ultrasound, MRI, and contrast-enhanced CT provides clarity and precision
for the diagnosis and extent of disease evaluation
of patients who have renal masses. Ultrasound
can be a highly useful adjunct in assessing cystic
lesions and allowing for a preoperative correlate
to intraoperative ultrasound, which is particularly
useful when a partial nephrectomy for an endophytic tumor is planned. MRI is the method of
choice for assessing the extent of renal vein and
inferior vena cava extension, which is essential
for assembling the proper surgical team and
equipment for such major operations. Unfortunately, none of these modalities can distinguish
with certainty a benign from malignant renal
mass or define the histologic subtype. A new molecular imaging strategy, termed immunoPET
scanning, is under active investigation and uses
the cG250, whose target, CA9, is abundant in the
malignant conventional clear cell carcinoma.
cG250 is linked to iodine-124, which has a 4-day
half-life and 23% positron emission, permitting adequate PET imaging. In a pilot study of 25 patients
who had renal masses imaged before resection,
the G250 scan accurately predicted the clear cell
histology in 13 of 14 cases with only one false negative. This imaging approach is currently being
evaluated in a large, multi-institutional national trial
of 158 patients. If these initial results are confirmed, the implications for the use of this clear
cellspecific immunoPET scan are broad and
could include a rational approach to active surveillance, molecular determination of extent of disease, extension of kidney-sparing operations,
and a molecular means of following the impact of
novel local treatments (renal tumor ablations) and
systemic agents (multitargeted kinases).
Preface
Surgeons managing renal cortical tumors confront two distinct patient groups. The first group,
approximately 30% of the total, is the large, usually symptomatic, and locally advanced tumors
often with regional adenopathy, adrenal invasion,
and extension into the renal vein or inferior venacava. Most of these tumors have the conventional clear cell histology (90%) and differ little
from the symptomatic tumors treated surgically
by Robson after World War II. Approximately
4% to 10% of patients have renal cancers that
involve the renal vein and inferior vena cava. In
1% of patients, the thrombus can extend into
the right atrium of the heart. Surgical resection
of these tumors can require complex cardiovascular techniques, with and without bypass, and
can lead to safe resection with perioperative mortality rates of less than 10% depending on the
extent of the thrombus. In the absence of nodal
or distant metastases, 5-year survival rates of
greater than 50% are possible. The incidence of
occult positive lymph nodes in 3% to 5% of patients has lead many surgeons to recommend
thorough retroperitoneal node dissections, especially for large T3 and T4 tumors, which may provide a therapeutic benefit in otherwise well
patients who have limited metastatic disease
and provide enhanced pathologic staging that
can facilitate entry into adjuvant clinical trials.
Despite radical resection of these massive tumors, whether by open or laparoscopic surgical
techniques, in conjunction with regional lymphadenectomy and adrenalectomy, progression to
distant metastasis and death from disease occurs
in approximately 30% of these patients. For patients presenting with or developing isolated metastatic disease with good performance status,
metastasectomy has been associated with longterm survival. For patients who have diffuse metastatic disease and good performance status,
cytoreductive nephrectomy may add several additional months of survival as opposed to cytokine
therapy alone. The integration of surgical therapy
for locally advanced and metastatic renal cancer
in the new era of effective systemic multitargeted
kinase inhibitors is currently under active investigation in neoadjuvant and adjuvant clinical
trials.
The second group, approximately 70% of the
total, is those who have small renal tumors (median tumor size <4cm, T1a), usually incidentally
discovered in asymptomatic patients during ultrasound, CT, or MRI obtained for other reasons.
A survival rate of 90% or greater, depending on
the tumor histology, is expected whether partial
or radical nephrectomy is performed. Approximately 20% of patients are found to have
xv
xvi
Preface
The remaining patients had vascular sclerotic
changes and other intrinsic renal abnormalities,
including diabetic nephropathy, glomerular hypertrophy, mesangial expansion, and diffuse
glomerulosclerosis.
Before the morbid traditional endpoint of endstage renal failure and dialysis (eGFR <15 mL/
min/1.73 m2), CKD (eGFR 1560 mL/min/1.73 m2)
is associated with increasing risk for cardiovascular events, hospitalization, and death, the likelihood of which increases as the eGFR decreases.
It is now estimated that there are 19 million adults
in the United States who have CKD. Unlike the
carefully selected and much younger kidney donors, renal tumor patients have a median age of
older than 60 years and often have common medical conditions, such as diabetes, hypertension,
and peripheral vascular disease, that can affect
baseline kidney function. Careful, prospective renal tumor tracking studies in patients initially not
treated surgically coupled with meta-analysis of
the published literature suggests that small renal
tumors have a slow yearly growth rate (approximately 0.3 cm/y). A substantial percentage of
small renal masses have benign or indolent histology (45%), and even if the patient has a conventional clear cell carcinoma, the likelihood of
metastatic disease for tumors of 4 cm or less is believed to be 5% to 7%. The clinical logic of offering
active surveillance to select patients who have
small renal masses with or without a confirmatory
biopsy, particularly those who are elderly or comorbidly ill, is gaining increasing acceptance particularly in light of concerns that overly aggressive
application of radical nephrectomy can worsen or
cause CKD. Despite this robust literature in favor
of kidney preservation strategies, evidence from
large cross-sectional national databases indicates
that RN is overused in the United States and
abroad and still accounts for 90% of kidney operations for small renal (T1) tumors. Overuse of RN is
a quality-of-care issue that needs to be carefully
addressed through educational programs and increased training in open and laparoscopic kidney-sparing operations. New approaches to the
small renal mass, including laparoscopic and robotic partial nephrectomy, and thermal ablative
approaches (radiofrequency ablation, cryoablation) have been actively pursued by committed clinician investigators in the United States and
abroad. Careful case selection and increased experience with the laparoscopic approach to partial
nephrectomy has rendered this a reasonable
alternative in skilled hands; however, concerns relating to the impact of warm ischemia on long-term
renal function and greater perioperative urologic
and non-urologic complications compared with
Preface
the above-described pathways to the same degree as the clear cell tumors and are therefore
less responsive to the newly FDA-approved
agents.
The authors of this issue of the Urologic Clinics
of North America provide a comprehensive review
of the great progress made, on numerous fronts,
in the understanding and management of renal
cortical tumors. Their work illuminates a clear
and hopeful path to our future goal of further
xvii
Dedic ation
paintings and artwork, which was very well
received.
As a youngster, I knew that my father was a busy
and respected physician. When I was older and
worked alongside him as a hospital orderly and
premedical student, I understood for the first
time the full dimensions and responsibilities of
his profession. Keeping current with his specialty
was extremely important to my father, and he
was an avid and committed reader of the literature
in his and other medical specialties. I was always
amazed at how knowledgeable and thoughtful he
was about the contemporary problems in urology
and urologic oncology. When I interviewed for residency positions I found that many academic urologists knew my father, who had referred patients
to them when their expertise represented the
best care.
His career spanned most of the twentieth century, from the discovery of antibiotics to the cloning of the human genome. Most importantly, C.
Paul Russo was a loving and dedicated parent to
my sisters and me, and a devoted spouse to my
mother. For this, especially, I will always be
grateful.
urologic.theclinics.com
Surgical Management
of Renal Tumors :
A Historical Perspective
Harry W. Herr, MD
KEYWORDS
FOUNDATIONS
The modern era of renal surgery began on August
2, 1869, just 139 years ago, when Gustav Simon
(18241876), then Professor of Surgery at Heidelberg, performed the first planned nephrectomy
on a living human being.1 His patient was a 46year-old woman named Margaretha Kleb, who
suffered from a pervious urinary fistula caused by
an injury to the ureter sustained during a prior laparotomy to remove an ovarian cyst. She survived
removal of her kidney and was permanently cured
of her fistula. Eighteen years later in 1887, Vincenz
Czerny (18421915), Simons successor at Heidelberg, performed the first partial nephrectomy to
remove a renal tumor.2 Both total and partial nephrectomy have become the hallmark surgical
procedures used today to treat renal tumors, and
their conception and evolution represent two of
the most important advances in medicine and
surgery.
Simon (Fig. 1) arrived at his decision to remove
a healthy, functioning kidney, plagued with many
questions for which he had no sure answers.
Would the remaining kidney supply the needs of
the body? Could the ligated renal artery withstand
the blood pressure of the aorta following absorption of the ligature? Could the initial hemorrhage
be controlled? Could wounds and infection of the
peritoneum be avoided? Still, there was historical
precedent for the operation.
For many years, physiologists had established
the fact that animals could survive after removal
of one kidney. In 1670, Zambecarri, along with
Rounhuysen, extirpated sound kidneys without
Department of Urology, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, 1275 York
Avenue, New York, NY 10021, USA
E-mail address: herrh@mskcc.org
Urol Clin N Am 35 (2008) 543549
doi:10.1016/j.ucl.2008.07.010
0094-0143/08/$ see front matter 2008 Elsevier Inc. All rights reserved.
urologic.theclinics.com
544
Herr
545
546
Herr
involving the kidney. The question is, whether such
a procedure is ever justifiable when the opposite
kidney is normal. I am inclined to think that in certain circumstances it may be.22 In 1948 he removed a 10-cm carcinoma from the left kidney of
a 52-year-old woman with a normal right kidney.
His decision was based on the pathologic studies
by Cahill (1948), showing that clear cell carcinomas arose from the cortex, were localized, surrounded by a capsule, grew by expansile growth,
rarely invaded surrounding structures, and generally spread by the bloodstream. He was also aware
that the autopsy studies by Bell and others (1938
1944) had revealed few metastases from small renal tumors. Bell23 reported that only 7% of tumors
less than 5 cm had metastasized, compared with
83% of tumors larger than 10 cm. In fact, small tumors rarely broke through the capsule and only
one isolated metastasis was noted among 38 tumors 3 cm or smaller in size. Microscopic studies
of tissue adjacent to tumors persuaded Vermooten that some tumors could be excised with only
a 1-cm margin without fear of local recurrence,
and local tumor excision should be attempted, especially in a solitary kidney or when there was
markedly impaired function of the opposite kidney.
Few urologists paid much attention to Vermooten or to the observations of Bell except to argue
that, because small tumors might metastasize,
that warranted total nephrectomy for all renal tumors, especially in cases with two kidneys. During
the next 40 years, partial nephrectomy was done
mostly for tumors in a solitary kidney, poor renal
function, or bilateral renal tumors. From 1950 to
1967, Zinman and Dowd collected only 18 essential cases of partial nephrectomy, adding 3 of their
own. At the same time, other progressive surgeons
reported individual cases of partial nephrectomy
for unilateral renal tumors when the other kidney
was considered satisfactory. Still, most urologists
believed that partial nephrectomy was unwarranted unless there was a compelling reason to
preserve renal function. Textbooks published between 1937 and 1970 do not mention partial
nephrectomy.24
LAPAROSCOPIC NEPHRECTOMY
Although Kelling was the first to try laparoscopy
using Nitzes cystoscope in 1901, laparoscopic
surgery evolved slowly and was largely limited to
the removal of small pieces of solid tissues (ovary,
lymph nodes) or excision of hollow organs (gallbladder). In 1991, Ralph Clayman took a laparoscopic leap forward when he and his team in St.
Louis successfully removed the right kidney and
a 3-cm tumor from an 85-year-old woman. The operative time was 7 hours and became the first operation where laparoscopic techniques were used
to remove a major solid organ from a human being.
Clayman knew he was breaking new ground. Like
Simon had done more than a century before, Clayman performed successive nephrectomies in pigs
to learn the laparoscopic operation and to perfect
a sack to contain the kidney in the abdomen. Engineers also devised an effective morcellator/evacuator to remove the specimen.34 Published as
a simple letter35, Claymans innovative and bold
first laparoscopic nephrectomy touched off urologys enduring courtship with laparoscopic surgery (Fig. 3).
Advances then progressed rapidly. The first partial nephrectomy went from bench to bedside in
less than a year. Seminal work done by Elspeth
McDougall and colleagues36 in the laboratory
was translated into clinical reality in 1993 when
Howard Winfield37, in Iowa, performed the first
laparoscopic partial nephrectomy. Still in the development stage today, laparoscopic partial
547
548
Herr
nephrectomy for renal tumors is a technically demanding procedure that has yet to recapitulate
all of the favorable features marking the successful
open operation, and its final place in the armamentarium of the surgical treatment of renal tumors
remains to be defined. Improvements are being
made regularly, however; and these, and robot
assisted surgery now widely available, are likely
to extend laparoscopic capabilities beyond
imagination.
REFERENCES
1. Simon G. Chirugie der Nieren. Stuttgart: Ferdinand
Enke, vol. II, p. 314, 1871.
35. Clayman RV, Kavoussi L, Soper NJ, et al. Laparoscopic nephrectomy. N Engl J Med 1991;324:
13701.
36. McDougall EM, Clayman RV, Chandhoke PJ, et al.
Laparoscopic partial nephrectomy in the pig model.
J Urol 1993;149:16336.
37. Winfield HN, Donovan JF, Clayman RV. Laparoscopic partial nephrectomy: initial case report for
benign disease. J Endourol 1993;7:5216.
38. Huang WC, Sevey AS, Russo P. Chronic kidney
disease after nephrectomy in patients with
renal cortical tumors. Lancet Oncol 2006;9:
73540.
39. Russo P, Goetzl M, Simmons R, et al. Partial nephrectomy: the rationale for expanding indications.
Ann Surg Oncol 2002;9:6807.
549
Pathologic Features
of Renal Cor tical
Tumors
Satish K.Tickoo, MD*, Anuradha Gopalan, MD
KEYWORDS
Renal cell carcinoma Oncocytoma Pathology
Morphologic features Immunohistochemistry
Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York
10065, USA
* Corresponding author.
E-mail address: tickoos@mskcc.org (S.K. Tickoo).
Urol Clin N Am 35 (2008) 551561
doi:10.1016/j.ucl.2008.07.001
0094-0143/08/$ see front matter 2008 Elsevier Inc. All rights reserved.
urologic.theclinics.com
Until the 1980s, renal cortical tumors had been traditionally classified as clear cell, granular cell, papillary and sarcomatoid carcinoma, or renal
oncocytoma. In 1985, Thoenes and colleagues1,2
first described chromophobe renal cell carcinoma
(RCC) in human beings, followed soon by its eosinophilic variant. This led to a reassessment of the
morphologic classification of renal tumors and
laid the framework for the clinicopathologically
more valid classification used today (Box 1). We
know now that granular cell carcinoma and
sarcomatoid carcinoma are not specific entities,
and that granular and sarcomatoid features can be
seen in a variety of renal tumors with markedly diverse biologic potential. The current histologic
classification has also been validated by a number
of molecular studies.35 The implications of this
now universally accepted classification system
are that publications about clinicopathologic aspects of renal tumors from the pre-1985 era may
not correspond to the current knowledge about
these tumors.
The better understanding of the molecular aspects of renal tumors in the recent past has resulted in the realization that differing molecular
pathways are involved in different renal tumors.
This has resulted in the development and usage
of multiple targeted therapies, particularly for advanced clear cell RCC, with promising initial results.6 At the same time, this knowledge has
provided pathologists with additional tools to investigate molecular pathway markers in different
subtypes of kidney tumors, and to potentially use
them for their differential diagnostic and
552
Box 1
Classification of adult renal cortical tumors
Benign
Renal oncocytoma
Papillary adenoma
Metanephric adenoma
Metanephric adenofibroma
Malignant
Clear Cell (conventional) RCC
Papillary RCC
Chromophobe RCC
Collecting duct carcinoma
Medullary carcinoma
Tubulocystic RCC
Acquired cystic disease of kidney-associated
RCC
RCC, unclassified
Mucinous tubular and spindle cell carcinoma
Translocation associated carcinomas
Tumors of indefinite malignant potential
Multilocular cystic RCC
Fig.1. Clear cell RCC. (A) The high-grade areas to the left have large, Fuhrman nuclear grade 4 cytology and nonclear (granular) cytoplasm. (HE;100). (B) Diffuse and membranous immunoreactivity for carbonic anhydrase IX
(CA IX) is characteristic (CA-IX; 100).
PAPILLARY RCC
Papillary RCC (PRCC) constitute up to 15% of all
renal cortical neoplasms. There are at least two
distinct types of papillary RCC, both at the morphologic and genetic level, and they appear to
have distinct clinical behaviors as well.2528 Architecturally, most of these tumors have a papillary,
tubular, or tubulo-papillary growth pattern. Some
have asolid growth pattern because of compression of the papillary structures, while others show
a glomeruloid appearance. The cytoplasm may
be amphophilic, eosinophilic, or even partially
clear. Papillary RCCs are often multifocal, are frequently associated with papillary adenomas (tumors less than 5 mm in size, by definition) and
are the most common RCC type with bilateral disease. Classically, papillary tumors display abundant lipid laden, foamy macrophages within
fibrovascular cores, a feature helpful in establishing the correct diagnosis. It needs to be emphasized that correct classification of these tumors
requires experience and a detailed review of all
available slides.
Some experts feel that the Fuhrman grading
scheme is well suited for papillary RCC but others
disagree.10,26,29,30 Delahunt and Eble25 have suggested a two-tiered system (types 1 and 2)
(Fig. 2) based on nuclear features and growth pattern characteristics, which has also been accepted in the latest World Health Organization
classification. Whether these tumors should be
graded and, if so, what system to use remains
controversial.
Cytogenetic and molecular studies have revealed distinct findings in PRCC that distinguishes
them from other renal epithelial tumors.31,32 The
majority of tumors, particularly type 1, are characterized by trisomy of chromosomes 7 and 17, as
well as loss of chromosome Y.3,31,3337 Approximately 10% of the sporadic PRCC also show somatic mutations in the c-MET gene, a genetic
abnormality that is common as a germline mutation in familial cases.38 Some investigators have
suggested that tumors exhibiting trisomy 7/17
only are likely to be benign, whereas those exhibiting additional genetic abnormalities will behave
aggressively, a hypothesis that has not been confirmed in the literature. Recently, a group from the
National Institutes of Health described mutations
in the fumarate hydratase gene associated with
a subset of type 2 papillary RCC.4 In most large
Fig. 2. Variants of papillary RCC. (A) Type 1, with low-grade cytology and abundant foamy histiocytes in the cores
of the papillae (HE; 100). (B) Type 2, with high-grade nuclei, eosinophilic cytoplasm, and nuclear pseudostratification (HE;100).
553
554
RENAL ONCOCYTOMA
CHROMOPHOBE RCC
Chromophobe RCC, first described in 1985 by
Thoenes and colleagues,1 constitute approximately 6% of renal cortical neoplasms. Because
of their cytoplasmic features, many of these tumors may previously have been classified as clear
cell or granular RCC. The morphologic features include a solid growth pattern with sheets of cells
Fig. 3. (A) Eosinophilic variant of chromophobe RCC showing nuclear irregularities and perinuclear clearing.
(B) Renal oncocytoma with abundant pink cytoplasm similar to that in (A), but uniform round nuclei and absence
of perinuclear halos (HE; 100).
TUBULOCYSTIC CARCINOMA
In the recent past, the morphologic spectrum of
collecting was expanded to include some lowgrade tumors characterized by a tubulocystic pattern of growth, tumor cells with low-grade nuclei,
and mucin production.64 More recently, the terminology of tubulocystic carcinoma of the kidney
has been proposed for these tumors.65,66 They
555
556
UNCLASSIFIED RCC
Unclassified RCC form up to 6% of all renal epithelial tumors,5,29 and include renal carcinomas that
do not fit into any of the usual subtypes of renal
cortical tumors.67 Thus, tumors of unrecognizable
cell or architectural types, or those with apparent
composites of the recognized types, are all included in this category. While many of the tumors
are of high cytomorphologic grade and aggressive
clinical behavior, by definition they are not a pure
entity and include many other low-grade and less
aggressive tumors.
It is expected that with accumulation of tumors
and with more experience gained with them, pathologists will be in a better position to recognize
tumors with similar clinicopathologic features
within this category. That ability would enable the
reclassification of groups of tumors from the unclassified category as distinct entities, which is
exemplified by the next few tumor types.
557
558
Table 1
Immunohistochemical profile of common renal cortical tumor subtypes using a small, practical panel
of antibodies
Antibody
Clear Cell
Papillary
Chromophobe
CA IX
11, Diffuse
membranous
CD10
11, Diffuse
membranous
, Focal,
predominantly
perinecrotic and
tips of papillae
1, Focal to diffuse,
usually luminal
membranous
11
CK7
Racemase
c-kit
11
REFERENCES
1. Thoenes W, Storkel S, Rumpelt HJ. Human chromophobe cell renal carcinoma. Virchows Arch B Cell
Pathol Incl Mol Pathol 1985;48:20717.
2. Thoenes W, Storkel S, Rumpelt HJ. Histopathology
and classification of renal cell tumors (adenomas,
oncocytomas and carcinomas). The basic cytological and histopathological elements and their use
for diagnostics. Pathol Res Pract 1986;181:12543.
3. Kovacs G, Wilkens L, Papp T, et al. Differentiation
between papillary and nonpapillary renal cell carcinomas by DNA analysis. J Natl Cancer Inst 1989;
81:52730.
Oncocytoma
, Focal cytoplasmic
11, Cytoplasmic
with peripheral
accentuation
, Focal cytoplasmic
11
, Focal cytoplasmic
1
4. Linehan WM, Walther MM, Zbar B. The genetic basis of cancer of the kidney. J Urol 2003;170:
216372.
5. Reuter VE, Tickoo SK. Adult renal tumors. In:
Mills Carter, Greenson Oberman, Reuter Stoler,
editors. Sternbergs diagnostic surgical pathology.
4th edition. Philadelphia: Lippincott Williams
& Wilkins; 2004. p. 195599.
6. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib
versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356:11524.
7. Bonsib SM. Renal veins and venous extension in
clear cell renal cell carcinoma. Mod Pathol 2007;
20:4453.
8. Bonsib SM, Gibson D, Mhoon M, et al. Renal sinus
involvement in renal cell carcinomas. Am J Surg
Pathol 2000;24:4518.
9. Thompson RH, Blute ML, Krambeck AE, et al. Patients with pT1 renal cell carcinoma who die from
disease after nephrectomy may have unrecognized
renal sinus fat invasion. Am J Surg Pathol 2007;31:
108993.
10. Reuter VE, Presti JC Jr. Contemporary approach to
the classification of renal epithelial tumors. Semin
Oncol 2000;27:12437.
11. Tickoo SK, Alden D, Olgac S, et al. Immunohistochemical expression of hypoxia inducible factor-1alpha and its downstream molecules in sarcomatoid
renal cell carcinoma. J Urol 2007;177:125863.
12. de Peralta-Venturina M, Moch H, Amin M, et al. Sarcomatoid differentiation in renal cell carcinoma:
a study of 101 cases. Am J Surg Pathol 2001;25:
27584.
13. Kovacs G, Erlandsson R, Boldog F, et al. Consistent
chromosome 3p deletion and loss of heterozygosity
in renal cell carcinoma. Proc Natl Acad Sci U S A
1988;85:15715.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
559
560
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
84. Pavlovich CP, Walther MM, Eyler RA, et al. Renal tumors in the Birt-Hogg-Dube syndrome. Am J Surg
Pathol 2002;26:154252.
85. Teh BT, Blennow E, Giraud S, et al. Bilateral multiple
renal oncocytomas and cysts associated with a constitutional translocation (8;9) (q24.1;q34.3) and
a rare constitutional VHL missense substitution.
Genes Chromosomes Cancer 1998;21:2604.
86. Al-Ahmadie HA, Alden D, Qin L-X, et al. Carbonic
anhydrase IX expression in clear cell renal cell
carcinoma: an immunohistochemical study comparing two antibodies. Am J Surg Pathol 2008;
32:37782.
87. Tu JJ, Chen Y-T, Hyjek E, et al. Carbonic anhydrase
IX as a highly sensitive and specific marker of clear
cell renal cell carcinoma: A comparative immunohistochemical study using a panel of commonly utilized
antibodies in the differential diagnosis of renal cell
tumors. Mod Pathol 2005;18:169A.
88. Al-Ahmadie HA, Alden D, Olgac S, et al. The role of
immunohistochemical evaluation of adult renal cortical tumors on core biopsy: an ex-vivo study. Mod
Pathol 2007;20:134A.
561
cancer syndromes with increased risk of associated kidney cancer also have been reported, and
it is likely that more may exist.
Improved techniques of genetic research have
facilitated the relatively recent discovery of the
mutations associated with many HRC syndromes.
Before identification of the genes responsible, heritable kidney cancer was identified based on the
constellation of clinical findings used to define
the phenotypic manifestations of the syndrome.
These findings still provide the best initial means
for evaluating individuals suspected of having
hereditary kidney cancer and for screening their
relatives.
Screening approaches have been described for
many of the known HRC syndromes based on the
study of families that have these diseases and the
ages at which relevant phenotypic expression may
be identified.3 Effective evaluation and treatment
is approached best with a multidisciplinary team
of physicians and nurses aware of the syndrome
and it management. Often, frequent testing and
imaging studies need to be coordinated between
services, as do medical and surgical interventions
when indicated. Combined approaches to care
may help limit the risks to patients associated
with the requirements of management, including
radiographic exposure and surgical procedures.
Genetic testing, although available and highly
reliable, must involve careful discussion of the
many factors and consequences that come into
play and should be performed by a trained genetic
counselor. Many patients are intensely interested
in pursuing genetic testing when there is a suspicion of a hereditary basis for their disease, but
Department of Surgery/Urology Division, Weill Cornell Medical College, Memorial Sloan Kettering Cancer
Center, Box 12, 1275 York Avenue, NY 10021, USA
E-mail address: colemaj1@mskcc.org
Urol Clin N Am 35 (2008) 563572
doi:10.1016/j.ucl.2008.07.014
0094-0143/08/$ see front matter 2008 Elsevier Inc. All rights reserved.
urologic.theclinics.com
564
Coleman
they may not be fully aware of the ethical and legal
issues that can be raised. These issues can include the duty to inform relatives, the impact on
spousal relationships within the family, and prenatal and infant testing.4 Many have raised concerns
about discrimination in employment or insurance
on the basis of a genetically inherited disease,
prompting the passage of the Genetic Information
Non-discrimination Act by the U.S. Congress in
2007. The expansion of the understanding of the
human genome and the search for genetic causes
for disease are promising but also are complex
and challenging for both the individual and society.
Investigations of patients who have clinical manifestations of HRC syndromes have helped identify
genes and associated germline mutations that
result in cancerous and benign neoplasms in
a variety of organ systems. Variable phenotypic
expression within families has been recognized,
although the mechanism remains unclear; it is
speculated that possible environmental influences
or epigenetic mechanisms are at play. Importantly,
the finding of cancer genes related to the formation of seemingly unrelated tumors in separate
organ systems has helped shed light on the pathogenesis of cancer in a variety of tissues and the
common pathways they share.
Although familial forms of kidney cancer affect
a limited group of patients, the investigation of
these syndromes has had valuable impact on the
management of renal cancer. For most major subtypes of renal cortical tumors there is an associated heritable form of the disease identified with
a familial cohort. Screening of affected families
and careful follow-up has provided a greater understanding of the natural history of these tumors,
allowing insight into growth and other characteristics of the different forms of kidney cancer.
Somatic mutations in these same genes have
been found responsible for a large percentage of
sporadic kidney cancers as well, forming the basis
for an emerging understanding of the molecular
pathogenesis for kidney cancer.
Table 1
Subtypes of von Hippel-Lindau (VHL) syndrome by phenotype
VHL Group
Phenotype
Genotype Mutations
Type
Type
Type
Type
Type
Deletions, truncations
Missense substitutions
1
2
2a
2b
2c
565
566
Coleman
Fig. 2. Operative images of multiple renal cysts and tumors before (left) and after (right) excision with nephronsparing techniques to treat all visible sites of disease.
Birt-Hogg-Dube Syndrome
Hornstein and Knickenberg35 were the first to
describe a syndrome of perifollicular skin lesions
567
568
Coleman
Hereditary Leiomyomatosis Renal
Cell Carcinoma
HLRCC is a rare inherited disorder in which
affected individuals are at risk of developing cutaneous and uterine leiomyomas and renal cell
carcinoma. Affected HLRCC kindreds are characterized by germline mutation of the Krebs cycle enzyme, fumarate hydratase (FH). Recent studies
have suggested that a poorly defined but particularly lethal form of renal cell carcinoma is associated with HLRCC. These tumors initially were
described histologically as papillary neoplasms
but, because of their aggressive clinical behavior,
were classified separately as type 2 papillary tumors or were potentially misclassified as collecting
duct tumors.53 With further experience and more
refined characterization of the pathologic features,
this nomenclature has been abandoned at experienced centers, and the term HLRCC renal tumors is used now.54
Genetics of hereditary leiomyomatosis renal
cell carcinoma
Genetic alteration in HLRCC has been mapped to
a region on chromosome 1 (1q42.3-43) containing
10 exons encoding for FH.55,56 Germline mutations
in the gene may be inherited in an autosomal dominant fashion. Phenotypic expression shows high
penetrance for cutaneous leiomyomas and uterine
fibromas, leading many early investigators to refer
to the syndrome as multiple cutaneous and uterine leiomyomatosis or multiple leiomyomatosis.55,57 The occurrence of renal tumors among
patients who have the syndrome has been estimated at 2% to 21%, a prevalence low enough
to prompt some researchers to continue to differentiate between multiple leiomyomatosis and
HLRCC although they represent the same disease.56,58,59 In the syndrome of HLRCC, loss of
heterozygosity reveals the tumor suppressor function of the gene, demonstrating impaired enzymatic function of FH with resultant effects in
Krebs cycle catabolism.60 Biallelic loss of FH in
several models indicate that aberrant signaling
may be mediated through HIF-dependent pathways, suggesting that mechanisms of pseudohypoxia may play a role in tumorigenesis in
a similar fashion to VHL.7,60,61 Reports of clear
cell histology with FH mutation in a patient who
had HLRCC may support a common mechanism
of tumorigenesis through HIF activation.62,63
Clinical features
Clinical features of this disease are notable for
uterine leiomyomas (fibroids) of significant severity
for which, in a recently described series in the
United States, as many as 89% of affected women
Tuberous Sclerosis
An autosomal dominant disorder with renal manifestations, tuberous sclerosis affects as many as
1 person in 6000. TSC typically is characterized
by seizures, mental retardation, and the development of hamartomas in multiple organs as a result
of mutations in either the TSC1 or TSC2 gene.69
The gene products hamartin and tuberin together
form a protein complex that inhibits activation of
the downstream pathways of mammalian target
of rapamycin.70 Aberrant function of the hamartin-tuberin complex through mutation reveals their
role as tumor suppressor genes.
TSC1 has been mapped to chromosome
9 (9q34), and TSC2 is found on chromosome
16 (16p13.3).7173 Similar to other heritable syndromes, TSC may express a spectrum of severity
depending on the type and location of the mutation. Differences in the phenotype have been
linked to mutations in either TSC1 or TSC2 with
more severe manifestations of the syndrome,
Table 2
Biologic basis of hereditary kidney tumor syndromes: comparison of described hereditary renal tumor
syndromes with biologic and clinical correlates
Syndrome
Gene/Protein
Function
Pathwaya
Phenotype
VHL
3p25/pVHL
Tumor
suppressor
HIF
HPRC
Oncogent
BHD
7q31/c-Met
tyrosine
kinase domain
17p12/folliculin
Hepatocyte
growth
factor /c-Met
Mammalian
target of
rapamycin
Central nervous
system/ocular
hemangioblastoma
Endolymphatic sac
tumors
Pheochromocytoma
Pancreatic
neuroendocrine
tumors
Clear cell renal cell
carcinoma
Papillary type 1 renal
cell carcinoma
HLRCC
1q42.3/fumarase
Tumor
suppressor
HIF
TSC
TSC1 9q34TSC2
16p13.3 tubulin/
hamartin
Tumor
suppressor
Mammalian
target of
rapamycin
Tumor
suppressor
Fibrofolliculomas
Pulmonary blebs
Chromophobe/
oncocytoma/clear
cell renal cell
carcinoma
Skin leiomyoma
Uterine leiomyoma/
sarcoma
HLRCC renal cancer
Angiomyolipoma
Clear cell renal cell
carcinoma
SUMMARY
REFERENCES
569
570
Coleman
2. Rini BI, Campbell SC, Rathmell WK. Renal cell carcinoma. Curr Opin Oncol 2006;18(3):28996.
3. Lonser RR, Glenn GM, Walther M, et al. von HippelLindau disease. Lancet 2003;361(9374):205967.
4. Offit K, Thom P. Ethical and legal aspects of cancer
genetic testing. Semin Oncol 2007;34(5):43543.
5. Molino D, Sepe J, Anastasio P, et al. The history of
von Hippel-Lindau disease. J Nephrol 2006;
19(Suppl 10):S11923.
6. Seizinger BR, Rouleau GA, Ozelius LJ, et al. Von
Hippel-Lindau disease maps to the region of chromosome 3 associated with renal cell carcinoma.
Nature 1988;332(6161):2689.
7. Isaacs JS, Jung YJ, Mole DR, et al. HIF overexpression
correlates with biallelic loss of fumarate hydratase in
renal cancer: novel role of fumarate in regulation of
HIF stability. Cancer Cell 2005;8(2):14353.
8. Pause A, Lee S, Worrell RA, et al. The von HippelLindau tumor-suppressor gene product forms a stable complex with human CUL-2, a member of the
Cdc53 family of proteins. Proc Natl Acad Sci U S A
1997;94(6):215661.
9. Lonergan KM, Iliopoulos O, Ohh M, et al. Regulation
of hypoxia-inducible mRNAs by the von HippelLindau tumor suppressor protein requires binding
to complexes containing elongins B/C and Cul2.
Mol Cell Biol 1998;18(2):73241.
10. Bratslavsky G, Sudarshan S, Neckers L, et al. Pseudohypoxic pathways in renal cell carcinoma. Clin
Cancer Res 2007;13(16):466771.
11. Linehan WM, Vasselli J, Srinivasan R, et al. Genetic
basis of cancer of the kidney: disease-specific
approaches to therapy. Clin Cancer Res 2004;
10(18 Pt 2):6282S9S.
12. Kaelin WG Jr. The von Hippel-Lindau tumor suppressor protein and clear cell renal carcinoma. Clin
Cancer Res 2007;13(2 Pt 2):680s4s.
13. Gallou C, Chauveau D, Richard S, et al. Genotypephenotype correlation in von Hippel-Lindau families
with renal lesions. Hum Mutat 2004;24(3):21524.
14. Ong KR, Woodward ER, Killick P, et al. Genotypephenotype correlations in von Hippel-Lindau
disease. Hum Mutat 2007;28(2):1439.
15. Wong WT, Agron E, Coleman HR, et al. Genotypephenotype correlation in von Hippel-Lindau disease
with retinal angiomatosis. Arch Ophthalmol 2007;
125(2):23945.
16. Chen F, Kishida T, Yao M, et al. Germline mutations
in the von Hippel-Lindau disease tumor suppressor
gene: correlations with phenotype. Hum Mutat
1995;5(1):6675.
17. Maranchie JK, Afonso A, Albert PS, et al. Solid renal
tumor severity in von Hippel Lindau disease is
related to germline deletion length and location.
Hum Mutat 2004;23(1):406.
18. Wong WT, Agron E, Coleman HR, et al. Clinical characterization of retinal capillary hemangioblastomas
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
57.
58.
59.
60.
61.
62.
571
572
Coleman
63. Sudarshan S, Linehan WM, Neckers L. HIF and
fumarate hydratase in renal cancer. Br J Cancer
2007;96(3):4037.
64. Toro JR, Nickerson ML, Wei MH, et al. Mutations in
the fumarate hydratase gene cause hereditary leiomyomatosis and renal cell cancer in families in North
America. Am J Hum Genet 2003;73(1):95106.
65. Launonen V, Vierimaa O, Kiuru M, et al. Inherited
susceptibility to uterine leiomyomas and renal cell
cancer. Proc Natl Acad Sci U S A 2001;98(6):
338792.
66. Refae MA, Wong N, Patenaude F, et al. Hereditary
leiomyomatosis and renal cell cancer: an unusual
and aggressive form of hereditary renal carcinoma.
Nat Clin Pract Oncol 2007;4(4):25661.
67. Grubb RL 3rd, Franks ME, Toro J, et al. Hereditary
leiomyomatosis and renal cell cancer: a syndrome
associated with an aggressive form of inherited
renal
cancer.
J
Urol
2007;177(6):20749
[discussion: 207980].
68. Sudarshan S, Pinto PA, Neckers L, et al. Mechanisms of disease: hereditary leiomyomatosis and
renal cell cancera distinct form of hereditary
kidney cancer. Nat Clin Pract Urol 2007;4(2):10410.
69. Al-Saleem T, Wessner LL, Scheithauer BW, et al.
Malignant tumors of the kidney, brain, and soft
tissues in children and young adults with the tuberous sclerosis complex. Cancer 1998;83(10):
220816.
Molecular Biology
of Renal Cor tical Tumors
Tobias Klatte, MD, Allan J. Pantuck, MD*
KEYWORDS
HIF VEGF Raf mTOR Therapy VHL
Department of Urology, Room B7-298 CHS, Box 951738, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA 90095-1738, USA
* Corresponding author.
E-mail address: apantuck@mednet.ucla.edu (A.J. Pantuck).
Urol Clin N Am 35 (2008) 573580
doi:10.1016/j.ucl.2008.07.006
0094-0143/08/$ see front matter 2008 Elsevier Inc. All rights reserved.
urologic.theclinics.com
Renal cell carcinoma (RCC) accounts for approximately 3% of all adult malignancies. The incidence
of RCC has been steadily increasing; in 2008, over
50,000 Americans will be newly diagnosed with
RCC and over 12,000 will die of the disease.1
The clinical management of RCC is rapidly evolving along with our understanding of the disease
process. Historically, RCC was regarded as
a single entity. Today, RCC is more accurately recognized as a family of cancers that results from
distinct genetic abnormalities that have unique
morphologic features but is uniformly derived
from renal tubular epithelium. The current World
Health Organization classification distinguishes
clear cell, papillary, chromophobe, collecting
duct, medullary, and unclassified RCC and other
rare entities.2 Advances in genetics and molecular
biology have provided insight into the genetic
alterations underlying the various renal cortical
tumor types and the subsequent downstream
molecular pathways involved in their tumorigenesis. These recognized differences reflect a greater
sophistication in tumor analysis based on cytology, histology, genetic aberrations, glycogen
content, electron microscopy of cytoplasmic
microvesicles, and immunohistochemistry of intermediate filament proteins. For example, the
discovery of the von Hippel-Lindau (VHL) tumor
suppressor gene and the hypoxia-induced pathway in clear cell RCC has provided a valuable
substrate for application of new strategies for
diagnosis, patient selection, and targeted therapy.3 This article briefly reviews the major genetic
alterations underlying our current understanding of
renal cortical tumors, addresses the crucial biologic pathways that become altered in the various
RCC subtypes (Fig. 1), and gives some
574
Fig.1. Crucial pathways involved in RCC biology and tumorigenesis (see further explanation in the text). CAIX/XII,
carbonic anhydrase IX/XII; CXCR4, the chemokine receptor fusin; 4E-BP, eukaryotic initiation factor 4E binding
protein; EGFR, epidermal growth factor receptor; eIF-4E, eukaryotic initiation factor 4E; HIF-1a, hypoxia-inducible
factor 1a; GLUT-1, glucose transporter 1; IGF, insulin-like growth factor; MAPK, mitogen-activated protein kinase;
mTOR, mammalian target of rapamycin; PDGF, platelet-derived growth factor; PI3K, phosphatidylinositol
3-kinase; PTEN, phosphatase and tensin homolog; TGF, transforming growth factor; VEGF, vascular endothelial
growth factor.
HYPOXIA-INDUCED PATHWAY
The hypoxia-inducible pathway plays a key role in
regulation of angiogenesis, glucose transport,
575
576
complex is disrupted when TSC2 is phosphorylated by Akt, relieving the Rheb GAP activity of
TSC2 and allowing Rheb to bind ATP. In the presence of ATP, Rheb switches GDP to GTP and subsequently activates mTOR.63 The major
downstream targets of mTOR are the translational
components ribosomal S6 kinase 1 (S6K1) and eukaryotic initiation factor 4E (eIF4E)-binding
protein 1 (4E-BP1). Activation of S6K1 enhances
the translation of mRNAs containing the 50-terminal oligopyrimidine tract, whereas phosphorylation
of 4E-BP allows the release of eIF4E that facilitates
mRNA binding to the 40S ribosomal subunit.64
Thus, mTOR stimulates the translation of proteins
required for the cell cycle progression from the G1
to the S phase.
PI3K signaling is terminated by degradation of
PIP3, which can be mediated by two different
phosphatases. SH2-containing inositol phosphatase dephosphorylates PIP3 at position D5 and
thereby produces phosphatidylinositol-3,4-biphosphat,65 whereas PTEN removes the
phosphate group at position D3, thereby converting PIP3 back to PIP2.66 PTEN function is
frequently impaired due to deletions or mutations,
resulting in constitutive activation of Akt and upregulation of the mTOR pathway.
RAS/RAF/MEK /EXTRACELLULAR
SIGNAL-REGULATED KINASE PATHWAY
The Ras/Raf/MEK/extracellular signal-regulated
kinase (ERK) signaling pathway is an important
mediator of tumor cell proliferation and angiogenesis. It plays a central role in regulating cell growth
by transmitting signals from tyrosine kinase receptors such as EGF receptor, HER-2, VEGF receptor,
PDGF receptor, and MET to the nucleus. Genes
encoding for proteins involved in this pathway
are frequently mutated in cancers.67 Aberrant signaling and activation of this pathway leads to
increased cell survival, cell cycle progression,
proliferation, angiogenesis, invasion and metastasis, inhibition of apoptosis, and resistance to radiation and chemotherapy.68
Signaling starts by binding of a ligand to one the
four erbB proteins, the most prominent of which is
the erbB1 (EGF receptor). After binding of the ligands, the receptor becomes phosphorylated on
tyrosine residues, allowing the Ras protein to bind
GTP and to become active. Receptor binding is followed by activation of Raf kinase, which phosphorylates and activates MEK, which phosphorylates
and activates ERK1/2, which phosphorylates
several substrates involved in many cellular responses, including translation and transcription of
important proteins such as S6 kinase.68,69
THERAPEUTIC TARGETS
Unraveling the molecular pathways underlying
RCC has permitted the design and development
of agents specifically targeting these pathways,
a process that is currently revolutionizing the medical management of RCC and making it a paradigm
for the targeted therapy of solid malignancies.
Numerous drugs are now available for the systemic therapy of RCC that interfere with certain
targets of the previously described pathways.
For example, sorafenib is a multikinase inhibitor
that was initially described as a Raf kinase inhibitor
but also targets VEGF receptor and PDGF receptor b. Sorafenib has been shown to be effective
in phase II and III trials and is now approved for
the treatment of advanced RCC.77,78 Another multityrosine kinase inhibitor, sunitinib, inhibits VEGF
receptor and PDGF receptor b, and showed a response rate of 31% in patients who had metastatic
RCC, leading to prolongation of progression-free
survival compared with interferon-a.79 The monoclonal anti-VEGF antibody bevacizumab also led
to improvement of progression-free survival.80,81
The mTOR inhibitor temsirolimus (CCI-779)
prolonged overall survival among patients who
had metastatic RCC and poor prognosis.82 Other
inhibitors of mTOR, such as everolimus
(RAD001), are currently being explored in clinical
SUMMARY
The last 10 years have witnessed a dramatic evolution in our understanding of RCC biology, which
has led to the development of novel medical therapies and revolutionized the approach to RCC
clinical management. This review considers the
genetic basis of RCC and the molecular mechanisms of the hypoxia-induced pathway, the
mTOR pathway, the ERK pathway, and the ubiquitin-proteasome pathway. All these molecular pathways are involved in RCC biology, tumorigenesis,
and progression, and serve as the source of new
rational treatment strategies based on the design
of small molecule inhibitors directed against their
targets.
REFERENCES
1. Jemal A, Siegel R, Ward E, et al. Cancer statistics.
CA Cancer J Clin 2008;58(2):7196.
2. Lopez-Beltran A, Scarpelli M, Montironi R, et al. 2004
WHO classification of the renal tumors of the adults.
Eur Urol 2006;49(5):798805.
3. Lam JS, Leppert JT, Figlin RA, et al. Role of molecular markers in the diagnosis and therapy of renal cell
carcinoma. Urology 2005;66(5 Suppl):19.
4. Cohen AJ, Li FP, Berg S, et al. Hereditary renal-cell
carcinoma associated with a chromosomal translocation. N Engl J Med 1979;301(11):5925.
5. Kovacs G, Brusa P, deRiese W. Tissue-specific
expression of a constitutional 3;6 translocation: development of multiple bilateral renal-cell carcinomas.
Int J Cancer 1989;43(3):4227.
6. Pathak S, Strong LC, Ferrell RE, et al. Familial renal
cell carcinoma with a 3;11 chromosome translocation limited to tumor cells. Science 1982;217(4563):
93941.
577
578
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
579
580
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75. Voges D, Zwickl P, Baumeister W. The 26S proteasome: a molecular machine designed for controlled
proteolysis. Annu Rev Biochem 1999;68:101568.
76. Glickman MH, Rubin DM, Coux O, et al. A subcomplex
of the proteasome regulatory particle required for ubiquitin-conjugate degradation and related to the COP9signalosome and eIF3. Cell 1998;94(5):61523.
77. Escudier B, Eisen T, Stadler WM, et al. Sorafenib in
advanced clear-cell renal-cell carcinoma. N Engl J
Med 2007;356(2):12534.
78. Ratain MJ, Eisen T, Stadler WM, et al. Phase II placebo-controlled randomized discontinuation trial of
sorafenib in patients with metastatic renal cell carcinoma. J Clin Oncol 2006;24(16):250512.
79. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib
versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356(2):11524.
80. Escudier B, Pluzanska A, Koralewski P, et al. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, doubleblind phase III trial. Lancet 2007;370(9605):210311.
81. Yang JC, Haworth L, Sherry RM, et al. A randomized
trial of bevacizumab, an anti-vascular endothelial
growth factor antibody, for metastatic renal cancer.
N Engl J Med 2003;349(5):42734.
82. Hudes G, Carducci M, Tomczak P, et al. Temsirolimus,
interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007;356(22):227181.
83. Bukowski RM, Kabbinavar FF, Figlin RA, et al. Randomized phase II study of erlotinib combined with
bevacizumab compared with bevacizumab alone
in metastatic renal cell cancer. J Clin Oncol 2007;
25(29):453641.
84. Bleumer I, Oosterwijk E, Oosterwijk-Wakka JC, et al.
A clinical trial with chimeric monoclonal antibody
WX-G250 and low dose interleukin-2 pulsing
scheme for advanced renal cell carcinoma. J Urol
2006;175(1):5762.
85. Ronnen EA, Kondagunta GV, Motzer RJ. Medullary
renal cell carcinoma and response to therapy with
bortezomib. J Clin Oncol 2006;24(9):e14.
Epidemiology, Clinical
Staging, and
Presentation of Renal
Cell Carcinoma
G. Joel DeCastro, MD, MPH*, James M. McKiernan, MD
KEYWORDS
Renal cell carcinoma Epidemiology
Department of Urology, Columbia University/New York Presbyterian Hospital, Atchley Pavillion, 11th floor,
161 Fort Washington Avenue, New York, NY 10032, USA
* Corresponding author.
E-mail address: gjd16@columbia.edu (G.J. DeCastro).
Urol Clin N Am 35 (2008) 581592
doi:10.1016/j.ucl.2008.07.005
0094-0143/08/$ see front matter 2008 Elsevier Inc. All rights reserved.
urologic.theclinics.com
582
CHANGES IN DEMOGRAPHICS
In addition to changes in disease characteristics at
presentation, significant changes have been noted
in the demographics of RCC. There continues to
be a strong gender preponderance, with men
accounting for two-thirds of RCC diagnoses and
deaths.15,16 Luciani and colleagues8 found that
the mean age of patients presenting with RCC
has increased from a mean of 57 years in 1982
to 63 years in 1997. When restricted to incidentally
detected tumors only, they found that mean age at
detection increased to 64 years. More recently, the
SEER cancer registry reported that median age at
diagnosis between 2000 and 2004 was 65 years.17
The causes for this shift in age at diagnosis are
unclear. It may in part be the result of increased
exposure to the health care system owing to
a greater number of comorbidities as a patient
ages.8
Race has also emerged as an important factor in
the epidemiology of RCC. Using the SEER cancer
registry, Vaishampayan and colleagues18 found
that between 1975 and 1998, incidence rates
among blacks increased by 4.5% compared with
2.9% in whites. Similarly, Chow and colleagues4
estimated the RCC incidence among white men
between 1975 and 1995 to be 2.3%, compared
with 3.9% in black men. During the same period
white women had a 3.1% annual increase, while
the rate in black women increased by 4.9% per
year.
The reason why blacks have had a higher
incidence rate of RCC during the past 2 decades
is uncertain. There is no evidence that RCC in
this population is being disproportionately
detected via increased use of abdominal imaging.
The underlying reason for this disparity is therefore
likely to involve changes in the affected population
itself.18 For example, it is possible that there is
increased exposure within this population to risk
factors associated with RCC. As described in
more detail later in this article, established risk
factors for RCC such as obesity and hypertension
affect the black population disproportionately.
MORTALITY RATES
Excellent 5-year survival rates have been reported
for the small, localized masses that comprise the
majority of newly diagnosed renal tumors today.
Tsui and colleagues found that incidentally
detected tumors were more likely to be smaller,
lower stage, and lower grade than symptomatic
lesions. Accordingly, 5-year disease-free survival
(DFS) rates were 85.3% and 62.5%, respectively.
583
584
RISK FACTORS
Smoking
There are myriad risk factors that have been linked
to the development of RCC.3032 Smoking,
obesity, and hypertension are the three most
well-established factors associated with RCC. A
recent review by Vineis and colleagues33 revealed
a relative risk of 1.5 to 2.0 for development of RCC
in persons who smoke 20 or more cigarettes daily.
Similarly, a cohort study of more than 350,000
Swedish men found a significantly increased risk
of RCC in individuals with any smoking history.34
More recently, a meta-analysis by Hunt and
colleagues35 in 2005 reviewed 19 case-control
studies and 5 cohort studies (Fig. 1). In their comparison of ever versus never smokers, they
found that the relative risk for RCC in smokers
was 1.39 (95% CI 1.271.5). Among men, the relative risk was 1.54 (1.421.68), and in women 1.22
(1.091.36). There was also a significant dose-dependent effect, with relative risk increasing in
those smoking more than 20 cigarettes per day
compared with fewer than 10. In men, the relative
Obesity
Obesity has long been considered an important
risk factor for developing RCC. In the previously
cited study, Chow and colleagues34 divided
patients into three groups based on increasing
BMI. They found that those in the middle group
had a 60% greater risk of developing RCC than
those in the lower group, while the risk in the highest BMI group doubled (P < .01). In a recent largescale study, 2 million Norwegian men and women
were followed for a mean of 23 years between
Fig.1. Forrest plot for estimated RR for ever smokers Q-test for heterogeneity (P 5 .083). (From Hunt JD, van der
Hel OL, McMillan GP, Boffetta P, Brennan P. Renal cell carcinoma in relation to cigarette smoking: meta-analysis of
24 studies. Int J Cancer 2005;114(1):1018; with permission.)
Hypertension
Multiple studies have demonstrated the association
between hypertension and RCC. Chow and colleagues34 noted a positive trend in RCC risk with increasing diastolic and systolic blood pressure. As
shown in Table 1, compared with individuals with diastolic blood pressures less than 70 mm Hg, those
with values higher than 90 mm Hg had a doubled
risk of developing RCC. Similarly, compared with
patients with a systolic blood pressure of less than
120 mm Hg, those with a value higher than
150 mm Hg had a 60% higher risk of RCC. In this
same study, no association was found between
blood pressure values and the risk of renal pelvis
CLINICAL STAGING
The goal of any staging system is to combine available data about malignant disease to make accurate prognoses, as well as to assist in choosing
appropriate treatment modalities and determine
eligibility of patients for clinical trials. For the past
3 decades the TNM (tumor-node-metastases)
staging system has been widely applied to RCC.
Instituted in 1974 by the International Union
Against Cancer47 and the American Joint Committee on Cancer (AJCC), the TNM system has since
undergone three major revisions, each time incorporating new data with the goal of improving its
prognostic accuracy.
In 1987, the size cut-off between T1 and T2 was
established as 2.5 cm, but was later demonstrated
to have poor prognostic value.48 In 1997, the size
cut-off for T2 disease was increased to 7 cm,
thereby improving the prognostic utility of the
system (Table 2).49,50
In 2002, the T1 stage was divided into T1a and
T1b substages, separated by a size cut-off of
4 cm.51 This change was largely prompted by the
increasing use of nephron-sparing surgery. Hafez
and colleagues52 examined 485 patients who
underwent partial nephrectomy, and found that
patients with tumors 4 cm or smaller in diameter
585
586
Table 1
Relative risk of RCC and renal-pelvic cancer among men according to smoking status, body mass index,
and blood pressure
Variable
No. Men in
Cohort
(n 5 362,992)
Smoking status
Nonsmoker
148,206
Former smoker
51,638
Current smoker 138,332
Unknown
25,816
Body mass index
%20.75
45,073
20.7521.90
45,131
21.9122.85
45,057
22.8623.80
46,516
23.8124.76
44,916
24.7725.95
45,987
25.9627.75
45,499
R27.76
45,813
P for trend
Follow-up
Renal-Cell Cancer
Renal-Pelvis Cancer
Person-Year
No. Men
with Cancer
Relative Risk
(95% CI)
No. Men
with Cancer
Relative Risk
(95% CI)
2,129,536
909,630
2,289,228
455,494
180
145
334
100
1.0
1.3
1.6
1.6
18
19
82
17
1.0 (ref)
1.6 (0.9 3.1)
3.5 (2.1 5.8)
2.6 (1.3 5.0)
705,242
707,289
710,225
741,832
720,615
744,218
735,804
718,663
32
46
43
78
107
102
156
195
1.0 (ref)
1.2 (0.7
0.9 (0.6
1.4 (0.9
1.6 (1.1
1.3 (0.8
1.7 (1.1
1.9 (1.3
<.001
1.8)
1.5)
2.1)
2.4)
1.9)
2.7)
2.7)
15
8
13
13
22
23
22
20
1.0 (ref)
0.4 (0.2
0.6 (0.3
0.5 (0.2
0.8 (0.4
0.7 (0.4
0.6 (0.3
0.5 (0.2
.25
1.0)
1.3)
1.1)
1.5)
1.5)
1.1)
1.0)
540,097
1,695,116
2,317,216
974,597
187,114
69,748
12
96
273
272
78
28
1.0 (ref)
1.4 (0.8
1.7 (0.9
2.1 (1.2
2.3 (1.2
2.2 (1.1
<.001
2.5)
3.0)
3.9)
4.4)
4.5)
6
22
49
46
10
3
1.0 (ref)
0.6 (0.2
0.6 (0.2
0.7 (0.3
0.6 (0.2
0.6 (0.1
.74
1.5)
1.4)
1.8)
1.9)
2.4)
554,138
1,561,807
1,754,399
1,122,128
458,505
332,911
28
103
192
168
124
144
1.0 (ref)
1.1 (0.7
1.5 (1.0
1.4 (0.9
1.6 (1.1
1.7 (1.1
.007
1.7)
2.2)
2.1)
2.4)
2.6)
11
28
29
27
20
21
1.0 (ref)
0.8 (0.4
0.6 (0.3
0.6 (0.3
0.7 (0.3
0.7 (0.3
.91
1.7)
1.2)
1.3)
1.5)
1.5)
(ref)
(1.0 1.6)
(1.3 1.9)
(1.2 2.0)
All models included age, smoking status, body-mass index (the weight in kilograms divided by the square of the height in
meters), and diastolic blood pressure. CI denotes confidence interval.
Data from Chow WH, Gridley G, Fraumeni JF Jr., Jarvholm B. Obesity, hypertension, and the risk of kidney cancer in men.
N Engl J Med 2000;343(18):130511.
Table 2
Revisions ofTNM staging for RCC
Extent of Disease
TNM 199749
TNM 200251
T3b
T3c
T4
T4a
Nx
N0
N1
Restricted to kidney, %7 cm
Restricted to kidney, %4 cm
Restricted to kidney, 4 cm7 cm
Restricted to kidney, >7 cm
Invading adrenal gland or perinephric
fat within Gerotas fascia
Extension into renal vein or IVC below
diaphragm
Extension into the IVC above the
diaphragm or invasion into its wall
Extension beyond Gerotas fascia
T1
T1a
T1b
T2
T3a
N2
N3
Metastases
Mx
M0
M1
IVC, inferior vena cava; RCC, renal cell carcinoma; TNM, tumor-node-metastases.
Nodal Status
TNM 198748
587
588
gland can often be visualized on preoperative imaging. It is therefore difficult to separate the controversies regarding the T3a substage into
clinical and pathologic arguments. Nevertheless,
the main controversy regarding the T3a substage
concerns the implied notion that extension into
perinephric and renal sinus fat represents a similar
risk level as adrenal invasion.48
A recent study from Columbia University demonstrated that T2 tumors had worse DFS than
those staged as pT3a, that is, tumor size larger
than 7 cm was a more important prognostic factor
than the presence or absence of perinephric fat invasion.64 With a median follow-up of 31 months,
DFS at 5 years for patients with T2 and T3a disease was 68% and 85%, respectively, leading to
the conclusion that tumor extension beyond the
renal capsule is not as important as tumor size.
Similarly, in a review of almost 1,800 patients
Siemer and colleagues65 found no independent
prognostic value for perinephric fat involvement.
Similar conclusions have been made by other
researchers.64 These findings call into question
whether there is any prognostic significance to
including perinephric fat invasion in a staging
system.
The inclusion of ipsilateral adrenal involvement
with the other features of T3a disease has also
been criticized.66 First, adrenal involvement in the
absence of extension beyond Gerotas fascia is
rare. In their prospective analysis of 128 nephrectomies for clinical stage T1 to T3b (N0M0) disease,
Kletscher and colleagues67 found that the incidence of ipsilateral adrenal involvement was less
than 2%. Other studies have echoed these
results.68
More important, however, is that adrenal
involvement appears to be associated with significantly worse outcome than perinephric or renal
sinus fat involvement.65 Han and colleagues69
noted that after radical nephrectomy, median
survival and 5-year DFS for patients with ipsilateral
adrenal gland involvement was 12.5 months and
0%, respectively, compared with 36 months and
36% for T3a patients with no adrenal involvement.
This difference held in multivariate analysis
controlling for grade and nodal status. Interestingly, they also found that the DFS of patients
with adrenal involvement was statistically equivalent to that for patients with T4 disease.
Tumor extension into the venous system is not
uncommon in newly diagnosed patients, with up
to 9% having involvement of either the renal vein
or vena cava.70 The 2002 TNM system separates
venous thrombus involvement into two categories:
T3b includes thrombus extending into the renal
vein and up to the inferior vena cava (IVC) below
SUMMARY
The increasing incidence of renal cell carcinoma
over the past 2 decades can be in part explained
by the expanding use of abdominal imaging. As
a result, the vast majority of incident renal cancers
today are small, localized, and asymptomatic.
However, the well-documented rise in all stages
of RCC, including advanced and metastatic disease, calls into question the nature of these
asymptomatic lesions. The expected screening
effect of detecting RCC when it is small and localized, with subsequent decreases in diseasespecific mortality, has not been observed. In fact,
disease-specific mortality is actually rising, especially in the black community.
The question of what these asymptomatic
lesions represent is important. Since effective
surgical therapies have not decreased overall
mortality rates from RCC, we must question our
present treatment paradigm of these incidentally
detected cancers. Evidence from available active
surveillance protocols provides some insight into
the natural course of these masses, but the
evidence is as of yet insufficient to establish clinical standards. As a result, the treatment of incidentally detected masses using aggressive,
extirpative therapies remains the norm, but with
more minimally invasive options as well as energy
ablative therapies being increasingly employed.
Racial disparities in the incidence of RCC may
be in part explained by the preponderance of
two well-established risk factors for RCC in the
black population. Multiple studies have shown
that both hypertension and obesity are indeed
associated with RCC, although the exact mechanism of the relationship has yet to be elucidated.
Blacks are particularly affected by these risk factors, which may help to explain the increasing incidence of RCC in this population. Effective
interventions aimed at reducing obesity, hypertension, and smokingespecially in the most vulnerable populationsmay help to reduce the
incidence of RCC in the future.
REFERENCES
1. Jemal A, Siegel R, Ward E, et al. Cancer statistics,
2007. CA Cancer J Clin 2007;57:4366.
2. Silverberg E, Holleb AI. Cancer statistics, 1971. CA
Cancer J Clin 1971;21:1331.
3. Hollingsworth JM, Miller DC, Daignault S, et al.
Rising incidence of small renal masses: a need to
reassess treatment effect. J Natl Cancer Inst 2006;
98(18):13314.
589
590
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
http://seer.cancer.gov/csr/1973_1996.
Accessed
February 1, 2008.
Davis CJJ, Mostofi FK, Sesterhenn IA. Renal medullary carcinoma. The seventh sickle cell nephropathy.
Am J Surg Pathol 1995;19(1):111.
Berndt SI, Carter HB, Schoenberg MP, et al. Disparities in treatment and outcome for renal cell cancer
among older black and white patients. J Clin Oncol
2007;25(24):358995.
Pantuck AJ, Zisman A, Belldegrun AS. The changing natural history of renal cell carcinoma. J Urol
2001;166(5):161123.
Parsons JK, Schoenberg MS, Carter HB. Incidental
renal tumors: casting doubt on the efficacy of early
intervention. Urology 2001;57(6):10135.
Welch HG, Schwartz LM, Woloshin S. Are increasing
5-year survival rates evidence of success against
cancer? JAMA 2000;284(16):29758.
Chawla SN, Crispen PL, Hanlon AL, et al. The natural history of observed enhancing renal masses:
meta-analysis and review of the world literature.
J Urol 2006;175(2):42531.
Schachter LR, Cookson MS, Chang SS, et al.
Second prize: frequency of benign renal cortical
tumors and histologic subtypes based on size in
a contemporary series: what to tell our patients.
J Endourol 2007;21(8):81923.
Frank I, Blute ML, Cheville JC, et al. Solid renal
tumors: an analysis of pathological features related
to tumor size. J Urol 2003;170(6 Pt 1):221720.
Duchene DA, Lotan Y, Cadeddu JA, et al. Histopathology of surgically managed renal tumors: analysis of
a contemporary series. Urology 2003;62(5):82730.
Collins S, McKiernan J, Landman J. Update on the
epidemiology and biology of renal cortical
neoplasms. J Endourol 2006;20(12):97585.
Lipworth L, Tarone RE, McLaughlin JK. The epidemiology of renal cell carcinoma. J Urol 2006;176(6):
23538.
Murai M, Oya M. Renal cell carcinoma: etiology, incidence and epidemiology. Curr Opin Urol 2004;
14(4):22933.
Vineis P, Alavanja M, Buffler P, et al. Tobacco and
cancer: recent epidemiological evidence. J Natl
Cancer Inst 2004;96(2):99106.
Chow WH, Gridley G, Fraumeni JF Jr, et al. Obesity,
hypertension, and the risk of kidney cancer in men.
N Engl J Med 2000;343(18):130511.
Hunt JD, van der Hel OL, McMillan GP, et al. Renal
cell carcinoma in relation to cigarette smoking:
meta-analysis of 24 studies. Int J Cancer 2005;
114(1):1018.
Parker AS, Cerhan JR, Janney CA, et al. Smoking
cessation and renal cell carcinoma. Ann Epidemiol
2003;13(4):24551.
Bjorge T, Tretli S, Engeland A. Relation of height and
body mass index to renal cell carcinoma in two
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54. Patard JJ, Shvarts O, Lam JS, et al. Safety and efficacy of partial nephrectomy for all T1 tumors based
on an international multicenter experience. J Urol
2004;171(6 Pt 1):21815.
55. Mitchell RE, Gilbert SM, Murphy AM, et al. Partial
nephrectomy and radical nephrectomy offer similar
cancer outcomes in renal cortical tumors 4 cm or
larger. Urology 2006;67(2):2604.
56. Ficarra V, Schips L, Guille` F, et al. Multiinstitutional
European validation of the 2002 TNM staging system in conventional and papillary localized renal
cell carcinoma. Cancer 2005;104(5):96874.
57. Salama ME, Guru K, Stricker H, et al. pT1 substaging in renal cell carcinoma: validation of the 2002
TNM staging modification of malignant renal epithelial tumors. J Urol 2005;173(5):14925.
58. Cheville JC, Blute ML, Zincke H, et al. Stage pT1
conventional (clear cell) renal cell carcinoma: pathological features associated with cancer specific survival. J Urol 2001;166(2):4536.
59. Ficarra V, Guille F, Schips L, et al. Proposal for revision of the TNM classification system for renal cell
carcinoma. Cancer 2005;104(10):211623.
60. Zisman A, Pantuck AJ, Chao D, et al. Reevaluation
of the 1997 TNM classification for renal cell carcinoma: T1 and T2 cutoff point at 4.5 rather than
7 cm better correlates with clinical outcome. J Urol
2001;166(1):548.
61. Bonsib SM. T2 clear cell renal cell carcinoma is
a rare entity: a study of 120 clear cell renal cell
carcinomas. J Urol 2005;174(4 Pt 1):1199202.
62. Klatte T, Patard JJ, Goel RH, et al. Prognostic impact
of tumor size on pT2 RCC: an international multicenter experience. J Urol 2007;178(1):3540.
63. Frank I, Blute ML, Leibovich BC, et al. pT2 Classification for renal cell carcinoma. Can its accuracy
be improved? J Urol 2005;173(2):3804.
64. Murphy AM, Gilbert SM, Katz AE, et al. Re-evaluation of the tumour-node-metastasis staging of locally
advanced renal cortical tumours: absolute size (T2)
is more significant than renal capsular invasion
(T3a). BJU Int 2005;95(1):2730.
65. Siemer S, Lehmann J, Loch A, et al. Current TNM
classification of renal cell carcinoma evaluated:
revising stage T3a. J Urol 2005;173(1):337.
66. Lam JS, Shvarts O, Leppert JT, et al. Renal cell carcinoma 2005: new frontiers in staging, prognostication and targeted molecular therapy. J Urol 2005;
173(6):185362.
67. Kletscher BA, Qian J, Bostwick DG, et al. Prospective analysis of the incidence of ipsilateral adrenal
metastasis in localized renal cell carcinoma. J Urol
1996;155(6):18446.
68. Tsui KH, Shvarts O, Barbaric Z, et al. Is adrenalectomy a necessary component of radical nephrectomy? UCLA experience with 511 radical
nephrectomies. J Urol 2000;163(2):43741.
591
592
Contemporar y
Radiologic Imaging
of Renal Cor tical Tumors
Ariadne M. Bach, MDa,*, Jingbo Zhang, MDb
KEYWORDS
Renal cortical tumor Renal cancer Renal cell carcinoma
Renal mass Kidney cancer Clear cell carcinoma
Papillary Chromophobe Oncocytoma
Magnetic resonance imaging (MR)
Computed tomography (CT) Ultrasound (US)
DETECTION
CT is considered the modality of choice for detection and diagnosis of renal cortical tumors. MR
imaging and ultrasonography (US) are problemsolving tools or are used in patients who have
contraindications to IV contrast. The improvement
a
Department of Radiology, Memorial Sloan Kettering Cancer Center, Rockefeller Outpatient Pavilion, 160 East
53rd Street, Radiology-8th floor, New York, NY 10022, USA
b
Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10021,
USA
* Corresponding author.
E-mail address: bacha@mskcc.org (A.M. Bach).
urologic.theclinics.com
594
Fig.1. A 55-year-old man who has prostate cancer and incidentally discovered renal angiomyolipoma indicated by
arrows. CT demonstrates the mass to have a HU of 59, consistent with fat. The mass is very echogenic on US.
presence of enhancement within the thrombus indicates tumor thrombus, whereas bland thrombus
would not enhance after contrast administration.
Solid renal cortical tumors
Enhancement is important in the characterization
of solid renal cortical tumor.
A study at our institution by one of the authors
(JZ) demonstrated that 90% of the clear cell cancers were hypervascular and heterogenous, 75%
were of the papillary type and were hypovascular,
and chromophobe often demonstrated moderate
enhancement. A mixed enhancement pattern
was most predictive of clear cell.38
Clear cell carcinoma tends to be hypervascular
and heterogeneous (Fig. 3).3842 Clear cell carcinomas demonstrate peritumoral vascularity more
frequently than other malignant renal tumors of
similar size.43
Papillary renal cell cancer is typically hypovascular and homogeneous (Fig. 4).38,42
The chromophobe is more variable in appearance. It may demonstrate moderate enhancement.38 The spoke-wheellike enhancement with
a central scar has been described as an important
imaging feature.44
Oncocytomas may overlap with renal cell carcinoma in imaging features and degree of enhancement.38,39,45 Classic angiographic findings for
oncocytoma are a spoke-wheel pattern, a homogeneous tumor blush, and a sharp, smooth rim.46
But none of these findings are specific and a renal
cell carcinoma may have any or all of the classic
findings.46 The diagnosis of oncocytoma may be
suggested if a central stellate scar is identified on
CT within an otherwise homogenous tumor.47
The presence of macroscopic fat is characteristic of an AML. AML may present without evidence
Fig. 2. A 57-year-old man who has 10 14 cm right renal clear cell invading the right renal vein and extending
into the IVC. (A) CT demonstrates mass and arrow indicates the thrombus. (B) MR image with arrows pointing out
the tumor thrombus in the renal vein and IVC.
595
596
Fig. 3. A 55-year-old man who has clear cell cancer. Renal protocol CT demonstrates an enhancing right partially
cystic mass with minimal calcification. The mass enhances from 16 (A, noncontrast CT) to 60 HU (B, postcontrast
CT).
the cystic renal mass is graded as Bosniak category 4 and considered malignant. The presence
of nodular or septal enhancement has been shown
to have the highest sensitivity for predicting malignancy with good to moderate interobserver
agreement.52
of macroscopic fatthe so-called AML with minimal fat.48,49 AMLs have been described as being
homogeneously high attenuation on the unenhanced study and demonstrated homogeneous
enhancement.48,49 Overlap has been reported
with renal cancer.38,48 Small (%3 cm) homogeneously enhancing renal masses on CT may be
an AML with minimal fat or renal cell carcinoma.50
Ultrasound
Fig. 4. A 64-year-old woman who has a papillary renal cell carcinoma in the upper pole of the left kidney. Cyst is
imaged in the upper pole of the right kidney. (A) Noncontrast CT. (B) Postcontrast CT.
MR Imaging
MR imaging has distinct advantages over other
imaging modalities in the detection and staging
of renal neoplasms, because of its intrinsic high
soft tissue contrast and direct multiplanar imaging
capabilities.61 In addition, pseudo-enhancement
artifacts that frequently afflict CT examinations
are typically not present on MR images. MR
Fig. 5. Spectrum of solid renal cortical tumors on US. (A) 5-cm hyperechoic oncocytoma. (B) Transverse US image of
the left kidney demonstrates a 1.9-cm hypoechoic papillary renal cell carcinoma indicated by calipers. (C) 5-cm
isoechoic right renal oncocytoma indicated by calipers.
597
598
Fig. 6. A 62-year-old man who has a 4.6-cm solid vascular clear cell carcinoma in the left upper pole on CT (A).
(B) US demonstrates flow in the mass on color Doppler.
Fig. 7. A 30-year-old woman who has right renal angiomyolipoma. (A) Axial T1-weighted in-phase. MR image
demonstrates a right renal mass containing areas of T1 hyperintensity. (B) Axial T1-weighted opposed-phase
MR image demonstrates drop-off of signal in the T1 hyperintense areas, indicating the presence of microscopic
fat. (C) Axial T1-weighted fat-saturated contrast-enhanced MR image demonstrates heterogeneous enhancement
in the right renal mass.
Fig. 8. A 58-year-old man who has right renal clear cell carcinoma. (A) Axial T2-weighted single-shot fast spin echo
MR image demonstrates a mildly T2-hyperintense mass in the right kidney. (B) Axial T1-weighted fat-saturated
contrast-enhanced MR image demonstrates heterogeneous enhancement in the renal mass.
599
600
STAGING
The sensitivity of CT for detection of regional
lymph node metastases is reported to be as high
as 95%.102 False-positive findings up to 58%
have been reported when a size criterion of 1.0
cm is used for determining nodal metastasis,
however.102
The most common metastatic sites from malignant renal cortical tumors are the lung, bone, brain,
liver, and mediastinum.103 With improved imaging,
renal tumors are being detected incidentally in
greater numbers at smaller sizes and earlier
stages, and distant metastases are unlikely in
these patients.
IMAGING FOLLOW-UP
At our institution the renal US is frequently the first
imaging obtained following renal surgery. CT of the
chest, abdomen, and pelvis is the modality of
choice for detection of local recurrence and
distant metastases. In patients allergic to iodinated contrast, gadolinium-enhanced MR imaging
of the abdomen and pelvis may be performed.
Recurrences occur in the lung, bone, nephrectomy
site, brain, liver, and the contralateral kidney.104
REFERENCES
1. Kovacs G, Akhtar M, Beckwith BJ, et al. The Heidelberg classification of renal cell tumours. J Pathol
1997;183(2):1313.
2. Beck SD, Patel MI, Snyder ME, et al. Effect of papillary and chromophobe cell type on disease-free
survival after nephrectomy for renal cell carcinoma.
Ann Surg Oncol 2004;11(1):717.
3. Eble JN, Bonsib SM. Extensively cystic renal neoplasms: cystic nephroma, cystic partially differentiated nephroblastoma, multilocular cystic renal cell
carcinoma, and cystic hamartoma of renal pelvis.
Semin Diagn Pathol 1998;15(1):220.
4. McKiernan J, Yossepowitch O, Kattan MW, et al.
Partial nephrectomy for renal cortical tumors: pathologic findings and impact on outcome. Urology
2002;60(6):10039.
5. Motzer RJ, Bacik J, Mariani T, et al. Treatment outcome and survival associated with metastatic renal
cell carcinoma of non-clear-cell histology. J Clin
Oncol 2002;20(9):237681.
6. Russo P. Renal cell carcinoma: presentation, staging, and surgical treatment. Semin Oncol 2000;
27(2):16076.
7. Russo P. Renal tumors: developing understanding
leads to developments in surgical treatment. BJU
Int 2006;97:910.
8. Smith SJ, Bosniak MA, Megibow AJ, et al. Renal
cell carcinoma: earlier discovery and increased
detection. Radiology 1989;170(3 Pt 1):699703.
9. Russo P. Localized renal cell carcinoma. Curr Treat
Options Oncol 2001;2(5):44755.
10. Warshauer DM, McCarthy SM, Street L, et al.
Detection of renal masses: sensitivities and specificities of excretory urography/linear tomography,
US, and CT. Radiology 1988;169(2):3635.
11. Jamis-Dow CA, Choyke PL, Jennings SB, et al.
Small (%3-cm) renal masses: detection with CT
versus US and pathologic correlation. Radiology
1996;198(3):7858.
12. Malaeb BS, Martin DJ, Littooy FN, et al. The utility of
screening renal ultrasonography: identifying renal
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
601
602
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
57.
603
604
95.
96.
97.
98.
99.
100.
101.
102.
103.
104.
Molecular Imaging
of Renal Cell Carcinoma
Rodolfo Perini, MD, Daniel Pryma, MD, Chaitanya Divgi, MD*
KEYWORDS
Molecular imaging PET Cancer phenotype
Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, University of
Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19105, USA
* Corresponding author.
E-mail address: chaitanya.divgi@uphs.upenn.edu (C. Divgi).
Urol Clin N Am 35 (2008) 605611
doi:10.1016/j.ucl.2008.07.015
0094-0143/08/$ see front matter 2008 Elsevier Inc. All rights reserved.
urologic.theclinics.com
606
Perini et al
expression of cell surface markers. PET cannot
distinguish between various positron-emitting
radioisotopes, and therefore only one PET tracer
can be studied at a given time. Most positronemitting radioisotopes have short half-lives, however, and thus serial studies are feasible.
Because PET essentially identifies molecular
features of cellular processes, it can be thought of
as the pre-eminent molecular imaging modality.
An overview of PET imaging of renal cancer follows.
GLUCOSE METABOLISM
FDG is the most frequently used PET radiotracer
and is the only one approved by the Food and
Drug Administration (FDA) for oncologic PET imaging.12 Like glucose, FDG is transported into cells
by glucose transporters and is phosphorylated
by hexokinase. It is not metabolized further, however, and thus it accumulates inside cells. Tumor
cells use glucose constitutively, as described by
Warburg,8 and hence demonstrate greater FDG
uptake, appearing as areas of increased FDG
accumulation.
FDG-PET is being used increasingly in staging
and response assessment in a variety of cancers,
such as lung, breast, lymphoma, melanoma, and
neoplasms of the gastrointestinal tract.12,13 Its role
in renal cancer is still controversial, however.14,15
FDG-PET reportedly has limited use in the
primary detection and staging of renal cell carcinoma. Because FDG is excreted primarily through
the urine, foci of increased uptake may be
mistaken as activity in the collecting system.
Hydration and diuretics potentially can be used
to avoid such confounding factors, as is done in
bladder cancer. In addition, hybrid imaging with
PET/CT scanners also has the potential to improve
the sensitivity of FDG-PET in renal tumors from the
levels initially reported.
Bachor and colleagues16 initially described
FDG-PET as having an overall sensitivity of 77%
in detecting the primary tumor. From a group of
26 patients who had pathologically proven renal
cell carcinomas, 20 were identified by FDG-PET;
the diagnostic accuracy depended on the degree
of tumor differentiation. Most importantly, FDGPET proved to be a very accurate method for
lymph node staging, with no false negatives in
this study.
Goldberg and colleagues17 used FDG-PET to
study patients who had renal tumors and indeterminate renal cysts. The authors found that in 9 of
10 patients FDG-PET depicted solid neoplasms
accurately. Except for one patient who had a renal
cyst and a 4-mm papillary neoplasm, all benign
lesions also were classified as such by FDG-PET.
ACETATE METABOLISM
Acetate is taken up by cells, converted into
acetyl-coenzyme A in the mitochondria, and then
Fig. 1. FDG-PET of a patient who has renal cell carcinoma. Although the tumor can be visualized clearly
(arrow), the intensity of uptake is much less than in
the collecting system in the same kidney.
TUMOR-ASSOCIATED ANTIGENS
Antigenic features of tumors are studied in both
hematologic and solid neoplasms to identify
features of the cancer phenotype. In solid tumors
this study usually is performed by immunohistochemistry.30 Increasingly, the standardization of
such approaches is seen as critical to their rational
use in treatment decision making.31 Immunohistochemistry is a snap-shot: it depicts the antigenic
distribution in a portion of a single tumor, and
only in tissue accessible to biopsy. It therefore is
useful but is not always representative of the antigenic composition of all tumors in the patient.
IODINE-LABELED cG250
cG250 is an antibody that is being evaluated as
a therapeutic agent in clear cell kidney cancer.
The antibody has been studied extensively in
clear cell renal cancer as a murine antibody35,36
and as a chimeric (Fv-grafted human IgG1) antibody.3537 The antibody had exquisite specificity
for clear cell renal cancer, and single-photon imaging with [131I]iodine demonstrated that targeting to both primary and metastatic tumor was
excellent.3537
The excellent and invariable targeting of radioiodinated cG250 to clear cell renal cancer led the
authors to postulate that the antibody could be
used to identify clear cell renal cancer. Additionally, the availability of I-124 and the ability to attach
it to cG250 raised the prospect of tumor characterization with PET.
As stated earlier,13 the demographics of renal
cancer have changed: an increasing proportion
of patients with renal masses now are identified
serendipitously, with a consequent decrease in
the proportion of patients who have the clear cell
607
608
Perini et al
Fig. 2. ImmunoPET with [124I]-cG250
(A) in a patient who has clear cell renal
cancer (uptake in the tumor is clearly
evident, as indicated by the arrow) and
(B) in a patient who has angiomyolipoma
(there is no uptake in the tumor).
cancer phenotype.38 The authors proposed a clinical trial to test the hypothesis that antibody PET
with [124I]-cG250 would detect clear cell renal
cancer with at least 90% accuracy. The Ludwig
Institute for Cancer Research sponsored the trial,
which was conducted solely at the Memorial
Sloan-Kettering Cancer Center under an
FDA-approved Investigational New Drug application. The demographics at the Memorial Sloan-Kettering Cancer Center39 led the authors to anticipate
a total accrual of 54 patients with an early analysis
planned after 15 PET-positive patients.
Patients who had renal masses scheduled for
surgical removal were entered into the study.
Fig. 3. ImmunoPET with [124I]-cG250.
(A) Transaxial CT, (B) transaxial [124I]cG250 PET, (C) transaxial fused PET-CT,
and (D) and coronal fused PET-CT images
of a 54-year-old man with right upper
pole clear cell renal cancer (arrows).
Images were acquired 5 days after intravenous infusion of 4 mCi 124I/10 mg
cG250 and immediately before right
upper pole partial nephrectomy.
Fig. 4. (A) Low-power digital autoradiograph from the clear cell renal cancer seen in Fig. 2. The circled area in
(A) is shown at high power as digital autoradiograph in (B) and as contiguous slice immunohistochemistry stained
with anti-CA-9 antibody in (C).
SUMMARY
Molecular imaging using PET permits in vivo
characterization of metabolic and cellular characteristics of cancers. The evaluation of renal cancer
using molecular imaging techniques has increased
significantly because of the need to characterize
REFERENCES
1. Israel GM, Bosniak MA. How I do it: evaluating renal
masses. Radiology 2005;236(2):44150.
2. Bosniak MA, Rofsky NM. Problems in the detection
and characterization of small renal masses. Radiology 1996;198(3):63841.
3. Raj GV, Thompson RH, Leibovich BC, et al. Preoperative nomogram predicting 12-year probability of
metastatic renal cancer. J Urol 2008;179(6):
214651 [discussion 2151].
4. Raj GV, Bach AM, Iasonos A, et al. Predicting the
histology of renal masses using preoperative Doppler ultrasonography. J Urol 2007;177(1):538.
5. Zhang J, Tehrani YM, Wang L, et al. Renal masses:
characterization
with
diffusion-weighted
MR
609
Perini et al
610
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
611
Prognostic Models
and Algorithms in Renal
Cell Carcinoma
Brian R. Lane, MD, PhDa, Michael W. Kattan, PhDb,*
KEYWORDS
Renal cell carcinoma Nomogram Prognostic algorithms
a
Glickman Urological and Kidney Institute, Cleveland Clinic, 9500 Euclid Avenue, A100, Cleveland, OH 44195,
USA
b
Department of Quantitative Health Sciences, Cleveland Clinic, 9500 Euclid Avenue, Wb-4, Cleveland, OH
44195, USA
* Corresponding author.
E-mail address: kattanm@ccf.org (M.W. Kattan).
urologic.theclinics.com
614
Box 1
Prognostic factors for RCC
Anatomic
Tumor size
PROGNOSTIC FACTORS
A criticism of staging systems is that they tend to
provide limited prognostic ability, in part because
there may be significant heterogeneity among
patients in each classification, and often the
systems do not take into account a number of
significant predictive factors.14 This criticism has
led investigators to explore various prognostic
factors, either alone or in combination with the
TNM staging system and/or other factors
(Box 1). Patients who present with either local or
systemic symptoms have a worse prognosis than
patients who have incidentally detected
tumors.1518 Moreover, the presence of symptoms
of cachexia, including weight loss, anorexia, or
malaise, or a reduction in overall health (Karnofsky
scale or Eastern Cooperative Oncology Group
[ECOG] performance status)19 at diagnosis
615
616
Reference
Motzer 1999
Mekhail 2005
Motzer 2004
Boumerhi 2003
Escudier 2007
Choueiri 2007
N
Previous treatment
670
None
353
None
137
Immunotherapy and
other therapies
92
85
Immunotherapy and
other therapies
85
300
Immunotherapy
93
120
VEGF-targeted
therapy
100
94
12.6
100
13.8a
Conventional
NA
histology (%)
Prior nephrectomy (%)
65
Median survival
10
(months)
Associated with adverse outcome?
Less than 1 to 2 years
Yes
from nephrectomy
or diagnosis to
metastasis
Hemoglobin below
Yes
lower limit of normal
Elevated alkaline
No
phosphatase
Abnormal corrected
Yes
calciumb
Yes
LDH more than
1.5 upper limit
of normal
Reduced performance
Yes
status
No. metastatic sites
No
Prior radiotherapy
No
Low platelet count
No
Low neutrophil count
No
85
81
14.8
74
12.7
85
16.5
Yes
No
No
Yes
Yes
Yes
Yes
Yes
No
No
No
No
Yes
Yes
No
Yes
Yes
Yes
Yes
Yes
Yes
No
No
Yes
No
No
Yes
No
No
Yes
Yes
Yes
No
No
No
No
No
No
No
No
No
No
Yes
No
No
No
No
No
Yes
Yes
Table 1
Prognostic factors used in prognostic algorithms for patients who have metastatic RCC
Fig. 1. Preoperative nomogram predicting the odds that an enhancing renal mass amenable to partial nephrectomy is a benign or malignant tumor. Instructions for physician: Locate the patients age on the age axis according to gender. Draw a line upwards to the points axis to determine how many points toward cancer the patient
receives for his (or her) age. Repeat this process for the other axes, each time drawing straight upward to the
points axis. Sum the points achieved for each predictor, and locate this sum on the total points axis. Draw
a straight line down to find the probability that cancer will be found after partial nephrectomy. Refer to this
probability as P(C) and find the probability of the tumor being benign, which is 1-P(C). Inform the patient:
Mr. (or Mrs.) X, if we had 100 men (or women) exactly like you, we would expect to find a kidney cancer in
P(C) of these patients after partial nephrectomy. (From Lane BR, Babineau D, Kattan MW, et al. A preoperative
prognostic nomogram for solid enhancing renal tumors 7 cm or less amenable to partial nephrectomy. J Urol
2007;178:431; with permission.)
617
618
Fig. 2. Postoperative nomogram predicting recurrence of conventional RCC after nephrectomy. Instructions for
physician: Locate the patients tumor size on the size axis. Draw a line upwards to the points axis to determine
how many points toward recurrence the patient receives for the tumor size. Repeat this process for the other
axes, each time drawing straight upward to the points axis. Sum the points achieved for each predictor and locate
this sum on the total points axis. Draw a straight line down to find the probability that the patient will remain
free of recurrence for 5 years, assuming the patient does not die of another cause first. Inform the patient:
Mr. (or Mrs.) X, if we had 100 men (or women) exactly like you, we would expect (predicted percentage from
nomogram) to remain free of their disease 5 years following surgery, although recurrence after 5 years is still possible. (From Sorbellini M, Kattan MW, Snyder ME, et al. A postoperative prognostic nomogram predicting recurrence for patients with conventional clear cell renal cell carcinoma. J Urol 2005;173:50; with permission.)
Table 2
Modified UCLA integrated staging system
Nonmetastatic cases
Stage
Fuhrman nuclear
grade
ECOG
performance status
Risk
Stage
Fuhrman nuclear
grade
ECOG
performance status
Risk
T1
T2
1-2
3-4
1
Low
Any
1
0
Low
Any
T4
2-4
2
1
Intermediate
Low
Any
High
3
1
Any
1
Intermediate
Metastatic cases
N2M0 or M1
N1M0
Any
T3
Any
4
1
Intermediate
1
High
Data from Zisman A, Pantuck AJ, Wieder J, et al. Risk group assessment and clinical outcome algorithm to predict the natural history of patients with surgically resected renal cell carcinoma. J Clin Oncol 2002:20;4561.
Table 3
SSIGN score algorithm
MOLECULAR FACTORS
Feature
T stage
pT1
pT2
pT3 or T4
N stage
pNx or N0
pN0 or N2
M stage
pM0
pM1
Tumor size
< 5 cm
R 5 cm
Furhman nuclear grade
1 or 2
3
4
Necrosis
Absent
Present
Score
0
1
2
0
2
0
4
0
2
0
1
3
0
2
619
620
Table 4
Estimated cancer-specific survival (CSS) following
radical nephrectomy for cRCC according to SSIGN
score
SSIGN
Score
1-Year
CSS (%)
5-Year
CSS (%)
10-Year
CSS (%)
01
2
3
4
5
6
7
8
9
10 or
more
100
99.1
97.4
95.4
91.1
87.0
80.3
65.1
60.5
36.2
99.4
94.8
87.8
79.1
65.4
54.0
41.0
23.6
19.6
7.4
97.1
85.3
77.9
66.2
50.0
38.8
28.1
12.7
14.8
4.6
Fig. 3. Postoperative nomogram predicting disease-specific survival based on clinical variables and molecular
markers (From Kim HL, Seligson D, Liu X, et al. Using protein expressions to predict survival in clear cell renal
carcinoma. Clin Cancer Res 2004;10:5468; with permission.)
SUMMARY
Patients should be provided with the most accurate information about their likely individual disease
course currently available to expert clinicians. A
preoperative nomogram can predict the likelihood
that a suspected malignancy is a cancer. Three
major prognostic algorithms exist for localized
RCC, each of which can assess the likelihood of
disease recurrence better than the TNM staging
system alone. Classification of patients who have
metastatic RCC into good-, intermediate-, and
poor-risk groups can define disease-specific survival better, aiding in the selection of appropriate
systemic therapies or clinical trials. In each of these
areas, more accurate nomograms will allow physicians to offer patients better counsel regarding
their likely clinical course, will assist in the planning
and tailoring of follow-up, and will identify patients
who are more likely to benefit from individual
treatments. Future algorithms predicting disease
outcomes or response to treatment are likely to
incorporate molecular factors with the potential to
provide even greater information.
REFERENCES
1. Janzen NK, Kim HL, Figlin RA, et al. Surveillance
after radical or partial nephrectomy for localized renal cell carcinoma and management of recurrent
disease. Urol Clin North Am 2003;30:84352.
2. Coppin C, Porzsolt F, Awa A, et al. Immunotherapy
for advanced renal cell cancer. Cochrane Database Syst Rev 2005;CD001425.
3. Chow WH, Devesa SS, Warren JL, et al. Rising
incidence of renal cell cancer in the United States.
JAMA 1999;281:162831.
4. Jemal A, Siegel R, Ward E, et al. Cancer statistics,
2007. CA Cancer J Clin 2007;57:4366.
5. Motzer RJ, Rini BI, Bukowski RM, et al. Sunitinib in
patients with metastatic renal cell carcinoma. JAMA
2006;295:251624.
6. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib
versus interferon alfa in metastatic renal-cell
carcinoma. N Engl J Med 2007;356:11524.
621
622
23. Kim HL, Belldegrun AS, Freitas DG, et al. Paraneoplastic signs and symptoms of renal cell carcinoma: implications for prognosis. J Urol 2003;
170:17426.
24. Kim HL, Han KR, Zisman A, et al. Cachexia-like
symptoms predict a worse prognosis in localized
T1 renal cell carcinoma. J Urol 2004;171:18103.
25. Ljungberg B, Grankvist K, Rasmuson T. Serum
acute phase reactants and prognosis in renal cell
carcinoma. Cancer 1995;76:14359.
26. Symbas NP, Townsend MF, El-Galley R, et al. Poor
prognosis associated with thrombocytosis in
patients with renal cell carcinoma. BJU Int 2000;
86:2037.
27. OKeefe SC, Marshall FF, Issa MM, et al. Thrombocytosis is associated with a significant increase in
the cancer specific death rate after radical
nephrectomy. J Urol 2002;168:137880.
28. Jacobsen J, Grankvist K, Rasmuson T, et al. Prognostic importance of serum vascular endothelial
growth factor in relation to platelet and leukocyte
counts in human renal cell carcinoma. Eur J Cancer
Prev 2002;11:24552.
29. Karakiewicz PI, Hutterer GC, Trinh QD, et al.
C-reactive protein is an informative predictor of
renal cell carcinoma-specific mortality: a European
study of 313 patients. Cancer 2007;110:12417.
30. Cheville JC, Lohse CM, Zincke H, et al. Comparisons of outcome and prognostic features among
histologic subtypes of renal cell carcinoma. Am
J Surg Pathol 2003;27:61224.
31. Lau WK, Cheville JC, Blute ML, et al. Prognostic
features of pathologic stage T1 renal cell carcinoma after radical nephrectomy. Urology 2002;59:
5327.
32. Moch H, Gasser T, Amin MB, et al. Prognostic utility
of the recently recommended histologic classification and revised TNM staging system of renal cell
carcinoma: a Swiss experience with 588 tumors.
Cancer 2000;89:60414.
33. Amin MB, Tamboli P, Javidan J, et al. Prognostic impact of histologic subtyping of adult renal epithelial
neoplasms: an experience of 405 cases. Am J Surg
Pathol 2002;26:28191.
34. Beck SD, Patel MI, Snyder ME, et al. Effect of papillary and chromophobe cell type on disease-free
survival after nephrectomy for renal cell carcinoma.
Ann Surg Oncol 2004;11:717.
35. Linehan WM, Pinto PA, Srinivasan R, et al. Identification of the genes for kidney cancer: opportunity
for disease-specific targeted therapeutics. Clin
Cancer Res 2007;13:671s9s.
36. Yao M, Yoshida M, Kishida T, et al. VHL tumor suppressor gene alterations associated with good
prognosis in sporadic clear-cell renal carcinoma.
J Natl Cancer Inst 2002;94:156975.
51.
52.
53.
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
nephron sparing surgery: importance of tumor histology and risk of metachronous recurrence. J Urol
2003;170:7603.
Dimarco DS, Lohse CM, Zincke H, et al. Long-term
survival of patients with unilateral sporadic multifocal renal cell carcinoma according to histologic
subtype compared with patients with solitary
tumors after radical nephrectomy. Urology 2004;
64:4627.
Lang H, Lindner V, Martin M, et al. Prognostic value
of multifocality on progression and survival in localized renal cell carcinoma. Eur Urol 2004;45:
74953.
Richstone L, Scherr DS, Reuter VR, et al. Multifocal
renal cortical tumors: frequency, associated clinicopathological features and impact on survival.
J Urol 2004;171:61520.
Hatcher PA, Anderson EE, Paulson DF, et al. Surgical
management and prognosis of renal cell carcinoma
invading the vena cava. J Urol 1991;145:203
[discussion: 234].
Kim HL, Zisman A, Han KR, et al. Prognostic significance of venous thrombus in renal cell carcinoma.
Are renal vein and inferior vena cava involvement
different? J Urol 2004;171:58891.
Ishimura T, Sakai I, Hara I, et al. Microscopic venous
invasion in renal cell carcinoma as a predictor of
recurrence after radical surgery. Int J Urol 2004;11:
2648.
Lang H, Lindner V, Letourneux H, et al. Prognostic
value of microscopic venous invasion in renal cell
carcinoma: long-term follow-up. Eur Urol 2004;46:
3315.
DallOglio MF, Antunes AA, Sarkis AS, et al. Microvascular tumour invasion in renal cell carcinoma:
the most important prognostic factor. BJU Int
2007;100:5525.
Lane BR, Babineau D, Kattan MW, et al. A preoperative prognostic nomogram for solid enhancing
renal tumors 7 cm or less amenable to partial
nephrectomy. J Urol 2007;178:42934.
Raj GV, Thompson RH, Leibovich BC, et al. Preoperative nomogram predicting 12-year probability of
metastatic renal cancer. J Urol 2008;179:214651.
Yaycioglu O, Rutman MP, Balasubramaniam M, et al.
Clinical and pathologic tumor size in renal cell carcinoma; difference, correlation, and analysis of the
influencing factors. Urology 2002;60:338.
Cindolo L, de la Taille A, Messina G, et al. A preoperative clinical prognostic model for non-metastatic
renal cell carcinoma. BJU Int 2003;92:9015.
Cindolo L, Patard JJ, Chiodini P, et al. Comparison
of predictive accuracy of four prognostic models
for nonmetastatic renal cell carcinoma after
nephrectomy. Cancer 2005;104:136271.
Sorbellini M, Kattan MW, Snyder ME, et al. A
postoperative prognostic nomogram predicting
623
624
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
77. Skubitz KM, Skubitz AP. Differential gene expression in renal-cell cancer. J Lab Clin Med 2002;
140:5264.
78. Huo L, Sugimura J, Tretiakova MS, et al. C-kit
expression in renal oncocytomas and chromophobe
renal cell carcinomas. Hum Pathol 2005;36:2628.
79. Junker K, Hindermann W, von Eggeling F, et al.
CD70: a new tumor specific biomarker for renal
cell carcinoma. J Urol 2005;173:21503.
80. Tretiakova MS, Sahoo S, Takahashi M, et al. Expression of alpha-methylacyl-CoA racemase in papillary renal cell carcinoma. Am J Surg Pathol 2004;
28:6976.
81. Yang XJ, Tan MH, Kim HL, et al. A molecular classification of papillary renal cell carcinoma. Cancer
Res 2005;65:562837.
82. Liu X, Minin V, Huang Y, et al. Statistical methods
for analyzing tissue microarray data. J Biopharm
Stat 2004;14:67185.
83. Kim HL, Seligson D, Liu X, et al. Using tumor
markers to predict the survival of patients with
metastatic renal cell carcinoma. J Urol 2005;173:
1496501.
84. Thompson RH, Gillett MD, Cheville JC, et al. Costimulatory B7-H1 in renal cell carcinoma patients:
indicator of tumor aggressiveness and potential
therapeutic target. Proc Natl Acad Sci U S A
2004;101:171749.
85. Krambeck AE, Dong H, Thompson RH, et al. Survivin and b7-h1 are collaborative predictors of
survival and represent potential therapeutic targets
for patients with renal cell carcinoma. Clin Cancer
Res 2007;13:174956.
86. Krambeck AE, Thompson RH, Dong H, et al. B7-H4
expression in renal cell carcinoma and tumor vasculature: associations with cancer progression
and survival. Proc Natl Acad Sci U S A 2006;103:
103916.
87. Thompson RH, Dong H, Lohse CM, et al. PD-1 is
expressed by tumor-infiltrating immune cells and
is associated with poor outcome for patients with
renal cell carcinoma. Clin Cancer Res 2007;13:
175761.
88. Seligson DB, Pantuck AJ, Liu X, et al. Epithelial cell
adhesion molecule (KSA) expression: pathobiology
and its role as an independent predictor of survival
in renal cell carcinoma. Clin Cancer Res 2004;10:
265969.
89. Parker A, Cheville JC, Lohse C, et al. Expression of
insulin-like growth factor I receptor and survival in
patients with clear cell renal cell carcinoma.
J Urol 2003;170:4204.
90. Rasmuson T, Grankvist K, Jacobsen J, et al. Serum
insulin-like growth factor-1 is an independent
91.
92.
93.
94.
95.
96.
97.
98.
99.
100.
101.
102.
103.
104.
105.
106.
107.
108.
109.
110.
111.
carcinoma using iodine-124-labelled antibody chimeric G250 (124I-cG250) and PET in patients
with renal masses: a phase I trial. Lancet Oncol
2007;8:30410.
Bui MH, Visapaa H, Seligson D, et al. Prognostic
value of carbonic anhydrase IX and KI67 as predictors of survival for renal clear cell carcinoma.
J Urol 2004;171:24616.
Atkins M, Regan M, McDermott D, et al. Carbonic
anhydrase IX expression predicts outcome of interleukin 2 therapy for renal cancer. Clin Cancer Res
2005;11:371421.
Cho D, Signoretti S, Dabora S, et al. Potential histologic and molecular predictors of response to temsirolimus in patients with advanced renal cell
carcinoma. Clin Genitourin Cancer 2007;5:37985.
Thompson RH, Kuntz SM, Leibovich BC, et al.
Tumor B7-H1 is associated with poor prognosis in
renal cell carcinoma patients with long-term
follow-up. Cancer Res 2006;66:33815.
Thompson RH, Dong H, Kwon ED. Implications of
B7-H1 expression in clear cell carcinoma of the
kidney for prognostication and therapy. Clin
Cancer Res 2007;13:709s15s.
Motzer RJ, Mazumdar M, Bacik J, et al. Survival
and prognostic stratification of 670 patients with
advanced renal cell carcinoma. J Clin Oncol
1999;17:253040.
Mekhail TM, Abou-Jawde RM, Boumerhi G, et al.
Validation and extension of the memorial sloan-kettering prognostic factors model for survival in
patients with previously untreated metastatic renal
cell carcinoma. J Clin Oncol 2005;23:83241.
Boumerhi G, Mekhail TM, Abou-Jawde RM, et al.
Prognostic factors for survival in previously treated
patients with metastatic renal cell cancer. J Clin
Oncol 2003;22 [abstract 1647].
Motzer RJ, Bacik J, Schwartz LH, et al. Prognostic
factors for survival in previously treated patients
with metastatic renal cell carcinoma. J Clin Oncol
2004;22:45463.
Escudier B, Choueiri TK, Oudard S, et al. Prognostic factors of metastatic renal cell carcinoma after
failure of immunotherapy: new paradigm from
a large phase III trial with shark cartilage extract
AE 941. J Urol 2007;178:19015.
625
a
Division of Urology, Department of Surgical Oncology, Princess Margaret Hospital and the University Health
Network, University of Toronto, 610 University Avenue, 3-124, Toronto, Ontario, Canada M5G 2C4
b
UroOncology Fellowship Program, University of Toronto, Toronto, Ontario, Canada
* Corresponding author.
E-mail address: m.jewett@utoronto.ca (M.A.S. Jewett).
urologic.theclinics.com
628
629
630
core is a safe procedure with a good accuracy.6567 The largest recent series with core and
FNA biopsy reported a low incidence of complication.6874 There are only six cases of tumor seeding in the clinical literature, so the overall
estimated risk is less than 0.01%.7581 There are
no recent reports of implantation; probably owing
to better technique including the use of guiding
cannulas.6567 Other complications such as infection, arteriovenous fistula, and pneumothorax are
extremely unlikely. Smith reported in 16,000 fine
needle biopsies an overall mortality rate of
0.031%.82 Maturen and colleagues66 studied the
accuracy of imaging-guided percutaneous renal
mass biopsy and its impact on clinical management. They performed 152 biopsies in 125 patients
with 97% malignancy sensitivity and 100% of
specificity. These results are supported by several
others reports showing a sensitivity and specificity
of 70% to 100% and 100%, respectively, but the
average sensitivity of FNA for cancer diagnosis is
lower (76% to 97%) than needle cores.64,8385
However, some report false-negative rates of up
to 24%.64 False-negative biopsies result from
sampling necrotic zones or missing the tumor entirely (which is easy to do, yielding normal kidney
or blood for example). These biopsies should be
classified as insufficient or unsatisfactory to
make a diagnosis to establish the incidence of
false-negative biopsies.
Lesion size is important. Rybicki and colleagues86 cautioned that negative results in percutaneous biopsies should be interpreted with
caution in masses smaller than 3 cm and larger
than 6 cm (10% of false-negative rates). Also, Lechevailler and colleagues71 showed that the rate of
biopsy failures increase in smaller lesions. Renal
masses smaller than 3 cm had a higher incidence
of biopsy failure compared with lesions larger than
3 cm (37% versus 9%). Technical changes by the
same group improved results.72
A good biopsy can establish tumor subtype and
grade, which may be helpful. Renshaw and colleagues87 studied 38 renal FNA specimens.
Seventy-four percent of the primary renal lesions
and all oncocytomas and the two chromophobe
tumors were correctly classified. With regard to tumor grade, Lechevallier and colleagues71 reported
a 74% concordance between surgical tissue and
percutaneous biopsy, with a 69% concordance
in lesions under 4 cm. However, other studies
have shown better correlation between pathological grade and the preoperative FNA (89% and
92% interobserver and intraobserver, variability).88
The use of percutaneous biopsies can have a remarkable effect on decision making and cost.
Wood and colleagues89 reported a 44% change
REFERENCES
1. Ries LAG, Melbert D, Krapcho M, et al, editors.
SEER cancer statistics review, 19752004. Bethesda
(MD): National Cancer Institute; 2007.
2. Karumanchi SA, Merchan J, Sukhatme VP. Renal
cancer: molecular mechanisms and newer therapeutic options. Curr Opin Nephrol Hypertens 2002;
11:3742.
3. Pantuck AJ, Zisman A, Belldegrun AS. The changing
natural history of renal cell carcinoma. J Urol 2001;
166:161123.
4. Hollingsworth JM, Miller DC, Daignault S, et al. Rising incidence of small renal masses: a need to reassess treatment effect. J Natl Cancer Inst 2006;98:
13314.
5. Chow WH, Devesa SS, Warren JL, et al. Rising incidence of renal cell cancer in the United States.
JAMA 1999;281:162831.
6. Hock LM, Lynch J, Balaji KC. Increasing incidence
of all stages of kidney cancer in the last 2 decades
in the United States: an analysis of surveillance, epidemiology and end results program data. J Urol
2002;167:5760.
7. Tsui KH, Shvarts O, Smith RB, et al. Renal cell carcinoma: prognostic significance of incidentally detected tumors. J Urol 2000;163:42630.
8. Rendon RA, Stanietzky N, Panzarella T, et al. The
natural history of small renal masses. J Urol 2000;
164:11437.
631
632
633
634
85. Niceforo J, Coughlin BF. Diagnosis of renal cell carcinoma: value of fine-needle aspiration cytology in
patients with metastases or contraindications to nephrectomy. AJR Am J Roentgenol 1993;161:13035.
86. Rybicki FJ, Shu KM, Cibas ES, et al. Percutaneous
biopsy of renal masses: sensitivity and negative predictive value stratified by clinical setting and size of
masses. AJR Am J Roentgenol 2003;180:12817.
87. Renshaw AA, Lee KR, Madge R, et al. Accuracy of
fine needle aspiration in distinguishing subtypes of
renal cell carcinoma. Acta Cytol 1997;41:98794.
88. Cajulis RS, Katz RL, Dekmezian R, et al. Fine needle aspiration biopsy of renal cell carcinoma. Cytologic parameters and their concordance with
histology and flow cytometric data. Acta Cytol
1993;37:36772.
89. Wood BJ, Khan MA, McGovern F, et al. Imaging
guided biopsy of renal masses: indications, accuracy and impact on clinical management. J Urol
1999;161:14704.
90. Silverman SG, Deuson TE, Kane N, et al. Percutaneous abdominal biopsy: cost-identification analysis.
Radiology 1998;206:42935.
91. Frank I, Blute ML, Cheville JC, et al. An outcome prediction model for patients with clear cell renal cell
carcinoma treated with radical nephrectomy based
on tumor stage, size, grade and necrosis: the SSIGN
score. J Urol 2002;168:2395400.
92. Walther MM, Choyke PL, Glenn G, et al. Renal cancer in families with hereditary renal cancer: prospective analysis of a tumor size threshold for
renal parenchymal sparing surgery. J Urol 1999;
161:14759.
93. Zisman A, Pantuck AJ, Chao D, et al. Reevaluation
of the 1997 TNM classification for renal cell carcinoma: T1 and T2 cutoff point at 4.5 rather than
7 cm better correlates with clinical outcome. J Urol
2001;166:548.
94. Mattar K, Basiuk J, Finelli A, et al. Active surveillance
of small renal masses: a prospective multi-centre
Canadian trial. Presented at the Annual EAU Congress, Milan, Italy. March 2629, 2008.
cytogenetic defects, and variable metastatic potentials ranging from the benign oncocytoma, to
the indolent papillary and chromophobe carcinomas, to the more malignant conventional clear
carcinoma.1 At our center, Memorial Sloan Kettering Cancer Center (MSKCC), the conventional
clear cell tumor accounts for 90% of all metastatic RCTs but only 54% of the renal tumors undergoing resection. Two groups of patients with
RCTs currently exist. The first group consists of
patients with symptomatic, large, locally advanced tumors often presenting with regional adenopathy, adrenal invasion, and extension into
the renal vein or inferior vena cava. Despite radical nephrectomy in conjunction with regional
lymphadenectomy and adrenalectomy, progression to distant metastasis and death from disease occurs in approximately 30% of these
patients. For patients presenting with isolated
metastatic disease, metastasectomy in carefully
selected patients has been associated with
long-term survival.2 For patients with diffuse metastatic disease and an acceptable performance
status, cytoreductive nephrectomy, compared
to cytokine therapy alone, may add several additional months of survival.3 Cytoreductive nephrectomy also prepares patients for integrated
treatment, now in neoadjuvant and adjuvant
a
Department of Surgery, Urology Service, Weill Cornell College of Medicine, Memorial Sloan Kettering Cancer
Center, 1275 York Avenue, New York, NY 10021, USA
b
Department of Urology, New York University School of Medicine, 150 East 32nd Street, Suite 200, New York,
NY 10016, USA
* Corresponding author.
E-mail address: russop@MSKCC.org (P. Russo).
urologic.theclinics.com
The medical and oncologic rationale for partial nephrectomy has evolved over the last 10 years and
is based on the following factors: an enhanced understanding of renal tumor histology, the proven oncological equivalency of partial and radical
nephrectomy for T1 renal cancers, and new concerns regarding chronic kidney disease (CKD) and
its potential adverse impact on cardiovascular health
and overall survival. Historically, partial nephrectomy
was reserved for patients with tumor in a solitary kidney, bilateral renal tumors, or tumor in a patient with
underlying medical diseases of the kidney or renal insufficiency. For the last 15 years, the concept of partial nephrectomy for patients with a renal tumor and
a normal contralateral kidney (kidney sparing or
nephron sparing), has generated increasing acceptance both in the United States and abroad, and,
over the last 5 years, has crystallized as the treatment of choice for small renal masses. In this article
we discuss the oncological and medical rationale for
partial nephrectomy as the treatment of choice
whenever possible for T1 (<7 cm) renal tumors.
636
637
638
639
640
resection to confirm the completeness of the ablation, it is not known whether ablation is as effective
as surgical resection and whether or not the radiological images postablation represent complete or
partial tumor destruction or simply a renal tumor,
partially treated, not in active growth. In a recent
report from the Cleveland Clinic, which has substantial experience in both radiofrequency ablation
and cryoablation, documented recurrence rates
for cryoablation were 13 of 175 cases (7.4%) and
26 of 104 cases (25%) for radiofrequency ablation
whose mean preablation tumor sizes were 3.0 and
2.8 cm respectively. Repeat ablations were performed in 26 patients but 12 patients were not candidates for repeat ablation because of large tumor
size, disease progression, or repeat ablation failure. Of these, 10 patients underwent attempted resection with only 2 patients being eligible for partial
nephrectomy (open) and 7 patients requiring radical nephrectomy. One operation was aborted.
From this data, it appears that a failed ablative procedure in a patient originally eligible for a partial
nephrectomy, likely translates into a radical nephrectomy as salvage procedure because of
extensive postablation scarring.54 Carefully
designed ablate and resect clinical protocols
need to be done, much like those done in the
1990s with cryotherapy and localized prostate
cancer, to determine the true effectiveness of
these approaches. For this same population of elderly patients or comorbidly ill patients with small
renal tumors, active surveillance is increasingly
being suggested as an alternative to invasive
treatments.55,56
SUMMARY
The value of partial nephrectomy in the management of small renal cortical tumors is gaining wider
recognition thanks to (1) enhanced understanding
of the biology of renal cortical tumors; (2) better
knowledge about tumor size and stage migration
to small tumors at the time of presentation; (3)
studies indicating the oncologic efficacy of kidney-sparing surgery, whether performed by open
or laparoscopic techniques, and (4) increasing
awareness of the wide prevalence of CKD and its
associated cardiovascular morbidity and mortality. The argument by many minimally invasive surgeons for laparoscopic radical nephrectomy and
its associated rapid convalescence and cosmesis
is not sufficiently compelling when iatrogenic initiation or worsening of CKD is the result. The overzealous use of radical nephrectomy for small renal
tumors, whether by open or laparoscopic techniques, must now be considered detrimental to
the long-term health and safety of the patient
641
642
REFERENCES
1. Linehan WM, Walther MM, Zbar B. The genetic basis
of cancer of the kidney. J Urol 2003;170:216372.
2. Russo P, Snyder M, Vickers A, et al. Cytoreductive
nephrectomy and nephrectomy/complete metastasectomy for renal cancer. The Scientific World Urology 2007;2:4252.
3. Flanigan RC, Salmon SE, Blumenstein BA, et al. Nephrectomy followed by interferon alfa-2b compared
with interferon alfa-2b alone for metastatic renalcell cancer. N Engl J Med 2001;345:16559.
4. Russo P. Renal cell carcinoma: presentation, staging, and surgical treatment. Semin Oncol 2000;27:
16076.
5. McKiernan JM, Yossepowitch O, Kattan MW, et al.
Partial nephrectomy for renal cortical tumors: pathological findings and impact on outcome. Urology
2002;60:10039.
6. Snyder ME, Bach A, Kattan MW, et al. Incidence of
benign lesions for clinically localized renal masses
<7cm in radiological diameter: influence of gender.
J Urol 2006;176:23916.
7. Dechet CB, Zincke H, Sebo TJ, et al. Prospective
analysis of computerized tomography and needle
biopsy with permanent sectioning to determine the
nature of solid renal masses in adults. J Urol 2003;
169:714.
8. Divgi C, Pandit-Taskar N, Jungbluth AA, et al. Preoperative characterization of clear cell renal carcinoma
using iodine-124 labeled antibody chimeric G250
(1241cG250) and positron emission tomography
(PET): phase 1 surgical validation in patients with renal masses. Lancet Oncol 2007;4:30410.
9. Russo P, Goetzl M, Simmons R, et al. Partial nephrectomy: the rationale for expanding the indications. Ann Surg Oncol 2002;9:6807.
10. Lee CT, Katz J, Shi WW, et al. Surgical management
of renal tumors of 4 cm or less in a contemporary
cohort. J Urol 2000;163:7306.
11. Leibovich BC, Blute ML, Cheville JC, et al. Nephron
sparing surgery for appropriately selected renal cell
carcinoma between 4 and 7 cm resulting in outcome
similar to radical nephrectomy. J Urol 2004;171:
106670.
12. Gilbert BR, Russo P, Zirinsky K, et al. Intraoperative
sonography. Application in renal cell carcinoma.
J Urol 1988;139:5824.
13. Stephanson A, Hakimian A, Snyder ME, et al. Complications of radical and partial nephrectomy in
a large contemporary cohort. J Urol 2004;171:
1304.
14. Yossepowitch O, Eggener SE, Serio A, et al. Temporary renal ischemia during nephron-sparing surgery
is associated with short-term but not long-term impairment of renal function. J Urol 2006;176(4 Pt 1):
133943.
15. Richstone L, Scherr DS, Reuter VR, et al. Multifocal
renal cortical tumors: frequency, associated clinicopathological features, and impact on survival. J Urol
2004;171:61520.
16. Tismit MO, Razin JP, Thionunn N, et al. Prospective
study of the safety margins in partial nephrectomy:
intraoperative assessment and contribution of frozen
section analysis. Urology 2006;67:9236.
17. Schachter LR, Bach AM, Snyder ME, et al. The
impact of tumor location on histological subtype of
renal cortical tumors. BJU Int 2006;98(1):636.
18. Yossepowitch O, Thompson RH, Leibovitch BC,
et al. Predictors and oncological outcomes following
positive surgical margins at partial nephrectomy.
J Urol 2008;179(6):215863.
19. Dash A, Vickers AJ, Schachter LR, et al. Comparison of outcomes in elective partial vs. radical
nephrectomy for clear cell renal cell carcinoma of
47 cm. BJU Int 2006;97:93945.
20. Pahernik S, Roos F, Rohrig B, et al. Elective nephron
sparing surgery for renal cell carcinoma larger than
4 cm. J Urol 2008;179:714.
21. Patel MI, Simmons R, Kattan MW, et al. Long term
follow up of bilateral sporadic renal tumors. Urology
2003;61:9215.
22. Kattan MW, Reuter V, Motzer RJ, et al. A postoperative prognostic nomogram for renal cell. J Urol 2001;
166:637.
23. Najaraian JS, Chavers BM, McHugh LE, et al. 20
years or more of follow-up of living kidney donors.
Lancet 1992;340:80710.
24. Fehrman-Ekholm I, Duner F, Brink B, et al. No evidence of loss of kidney function in living kidney
donors from cross sectional follow up. Transplantation 2001;72:4449.
25. Goldfarb DA, Matin SF, Braun WE, et al. Renal outcome 25 years after donor nephrectomy. J Urol
2001;166:20437.
26. Kaplan C, Pasternack B, Shah H, et al. Age-related
incidence of sclerotic glomeruli in human kidneys.
Am J Pathol 1975;80:22734.
27. Bijol V, Mendez GP, Hurwitz S, et al. Evaluation of the
nonneoplastic pathology in tumor nephrectomy
specimens: predicting the risk of progressive renal
failure. Am J Surg Pathol 2006;30:57584.
28. Coresh J, Selvin E, Stevens LA, et al. Prevalence of
chronic kidney disease in the United States. JAMA
2007;298:203847.
29. Stevens LA, Coresh J, Green T, et al. Assessing
kidney functionmeasured and estimated glomerular filtration rate. N Engl J Med 2006;354:
247383.
41. Sorbellini M, Kattan MW, Snyder ME, et al. Prognostic nomogram for renal insufficiency after radical or
partial nephrectomy. J Urol 2006;176:4726.
42. Huang WC, Levey AS, Serio AM, et al. Chronic kidney disease after nephrectomy in patients with renal
cortical tumors: a retrospective cohort study. Lancet
Oncol 2006;7:73540.
43. Thompson HR, Boorjian SA, Lohse CM, et al. Radical nephrectomy for pT1a renal masses may be associated with decreased overall survival compared
to partial nephrectomy. J Urol 2008;179:46873.
44. Jemal A, Siegel R, Ward E, et al. Cancer statistics,
2008. CA Cancer J Clin 2008;58:7196.
45. Hollenback BK, Tash DA, Miller DC, et al. National
utilization trends of partial nephrectomy for renal
cell carcinoma: a case of underutilization? Urology
2006;67:2549.
46. Miller DC, Hollingsworth JM, Hafez KS, et al. Partial
nephrectomy for small renal masses. An emerging
quality of care concern? J Urol 2006;175:8537.
47. Nuttail M, Cathcart P, van der Meulen J, et al. A description of radical nephrectomy practice and outcomes in
England. 19952002. BJU Int 2005;96:5861.
48. Scherr DS, Ng C, Munver R, et al. Practice patterns
among urologic surgeons treating localized renal
cell carcinoma in the laparoscopic age: technology
vs. oncology. Urology 2003;62:100711.
49. Ramani AP, Desai MM, Steinberg AP, et al. Complications of laparoscopic partial nephrectomy in 200
cases. J Urol 2005;173:427.
50. Kim FJ, Rha KH, Hernandez F, et al. Laparoscopic
radical versus partial nephrectomy: assessment of
complications. J Urol 2003;170:40811.
51. Gill IS, Kavoussi LR, Lane BR, et al. Comparison of
1800 laparoscopic and open partial nephrectomies
for single renal tumors. J Urol 2007;178:416.
52. Diblasio CJ, Snyder ME, Russo P. Mini flank supraeleventh incision for open partial or radical nephrectomy. BJU Int 2006;97(1):14956.
53. Gill IS, Remer EM, Hasan WA, et al. Renal cryoablation: outcome at 3 years. J Urol 2005;173:19037.
54. Nguyen CT, Lane BR, Kaouk JH, et al. Surgical salvage of renal cell carcinoma recurrence after thermal ablative therapy. J Urol 2008;180:1049.
55. Wehle MJ, Thiel DD, Petrou SP, et al. Conservative
management of incidental contrast-enhancing renal
masses as safe alternative to invasive therapy. Urology 2004;64:49.
56. Volpe A, Jewett MA. The role of surveillance for small
renal masses. Nat Clin Pract Urol 2007;4:23.
643
Choice of Operation
for Clinically Localized
Renal Tumor
CarvellT. Nguyen, MD, PhDa, Steven C. Campbell, MD, PhDb,
Andrew C. Novick, MDa,*
KEYWORDS
Renal cell carcinoma Radical nephrectomy
Partial nephrectomy Laparoscopy Thermal ablation
Intensive study of the biology of renal cell carcinoma (RCC) has advanced the understanding of
its pathogenesis and led to novel adjuvant therapies, such as tyrosine kinase inhibitors. Even in
this era of molecular targeted therapy, however,
surgical excision remains the primary curative
treatment for RCC. Historically, radical nephrectomy (RN), with or without ipsilateral adrenalectomy and regional lymphadenectomy, has been
the reference standard for curative treatment of
localized RCC. With prevailing data showing
excellent oncologic efficacy, RN long has been
the treatment of choice in patients who have localized unilateral RCC and a normally functioning
contralateral kidney.1
With the advent of laparoscopy, minimally invasive techniques have been applied to RN resulting
in an appealing alternative to open surgery that is
associated with decreased morbidity and quicker
convalescence. Generally used in the management of localized RCC without invasive features
or substantial venous or nodal involvement, these
techniques have had survival outcomes comparable with those of the open approach, and laparoscopic RN now is firmly established as one of the
standards in this field.25
Despite its long history and proven efficacy
in treating kidney cancer, changes in the
a
Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue A100, Cleveland,
OH 44195, USA
b
Section of Urological Oncology, Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, 9500
Euclid Avenue A100, Cleveland, OH 44195, USA
* Corresponding author.
E-mail address: novicka@ccf.org (A.C. Novick).
urologic.theclinics.com
646
Nguyen et al
a decreased incidence of chronic kidney disease
(defined by a serum creatinine > 2 mg/dL) as well
as a decreased rate of proteinuria at a follow-up
of 10 years. Other groups have demonstrated similar findings, and the increased incidence of renal
insufficiency following RN persists even after controlling for potential confounding factors such as
diabetes or hypertension.10,11
The primary concern from these studies is that
RN can have deleterious effects on a patients
long-term renal function and overall health,
because population-based studies have shown
a correlation between chronic kidney disease
and increased risks of morbid cardiac events, hospitalization for any reason, and death. Another
concern with indiscriminate application of RN is
the possibility of late recurrence of RCC in the
contralateral kidney; options for salvage treatment
become more limited in the context of a solitary
kidney. Overall, the current evidence supports
a judicious use of RN in the treatment of localized
renal masses that otherwise may be amenable to
NSS, and this principle now is an overriding
consideration in the management of patients who
have small renal masses. Undoubtedly, RN
remains a viable option when the primary tumor
is prohibitively large (ie, > 7 cm), replaces too
much renal parenchyma, is in a location not amenable to NSS, or in some older patients who desire
surgical excision but for whom the risks associated with a complex PN may not be acceptable.
Table 1
Outcomes after elective partial nephrectomy for unilateral renal tumor and normal opposite kidney
Series
No. of
Patients
Disease-Specific
Survival (%)
Local
Recurrence (%)
MeanTumor
Size (cm)
Bazeed et al (1986)
Carini et al (1988)
Morgan and Zincke (1990)
Selli et al (1991)
Provet et al (1991)
Steinbach et al (1992)
Moll et al (1993)
Thrasher et al (1994)
Lerner et al (1996)
DArmiento et al (1997)
van Poppel et al (1998)
Herr (1999)
Hefez et al (1999)
Barbalias et al (1999)
Belldegrun et al (1999)
Filipas et al (2000)
Delakas et al (2002)
Total
23
10
20
20
19
72
98
6
54
19
51
70
45
41
63
180
118
909
100
90
100
90
100
94.4
100
92
96
98
97.5
100
97.5
100
98
97.3
90100
0
0
0
0
0
2.7 (2 cases)
1 (1 cases)
0
5.6 (3 cases)
0
0
1.5 (2 cases)
0
7.3 (3 cases)
3.2 (2 cases)
1.6 (3 cases)
3.9 (cases)
07.3
3.3
3.5
3.1
< 3.5
2.6
N/A
4
4.3
<4
3.34
3
3
<4
3.5
<4
3.3
3.4
24.3 cm
RCC demonstrated a significantly lower rate of recurrence and improved survival for tumors smaller
than 4 cm.24 Such data provided the rationale for
using a 4-cm cutoff as the upper limit for elective
PN and a revision of the American Joint Committee on Cancer staging system for confined RCC
in 2002. During the last decade, PN has indeed
become a standard of care for the treatment of
T1a tumors. With the excellent oncologic efficacy
displayed by PN and the importance of renal preservation, some institutions now are expanding the
indications for elective PN to include T1b tumors
(47 cm) (Fig. 1).
Several studies have suggested that elective PN
in carefully selected patients can achieve oncologic efficacy equivalent to RN in the treatment of
stage T1b renal masses.3133 For example, a large
multi-institutional study demonstrated no significant difference in the rates of distant or local recurrence or cancer-specific mortality between
patients undergoing PN and those undergoing
RN for T1b tumors.31 Similarly, Leibovich and colleagues32 found comparable cancer-specific and
distant metastases-free survival rates for patients
treated with either PN or RN for pT1b tumors after
controlling for pathologic features such as stage,
647
648
Nguyen et al
Fig.1. A 50-year-old man who had no significant past medical history presented with microscopic hematuria and
was found to have a 4.8-cm left renal mass abutting the renal vein and branches of the renal artery (A), The contralateral kidney was normal. Despite the precarious hilar location, three-dimensional reconstructed images
(B) revealed a potential plane of dissection between the mass and the hilar vessels. OPN was performed, resulting
in complete excision of the tumor with negative margins while preserving 75% of the left kidney. Final pathology
was grade 3, pT2 clear cell RCC. At 2-year follow-up, the patient is without evidence of recurrence and has normal
renal function (serum creatinine, 1.0 mg/dL).
function, and postoperative morbidity and provides useful information about the relative merits
of these approaches.37 This study comprised
1800 patients including 771 undergoing LPN and
1029 undergoing OPN for a single renal tumor
smaller than 7 cm. Similar cancer-specific survival
(99.3% versus 99.2% at 3 years) and postoperative renal function (97.9% versus 99.6% functioning renal units at 3 months) was demonstrated.
Compared with OPN, LPN was associated with
decreased operative time, blood loss, and hospital
stay but demonstrated longer warm ischemia
times and more postoperative complications requiring additional interventions (Table 2).37 In particular, urologic complications (primarily urine leak
and hemorrhage) were more common after LPN
(9.2% versus 5.0%, P 5 .0006), and postoperative
hemorrhage was almost threefold more common
after LPN (4.2% versus 1.6%, P 5 .0002). It should
be noted that patients treated with OPN were
a higher-risk group than the LPN cohort: a greater
percentage of patients in the OPN group demonstrated decreased performance status, impaired
renal function, and symptomatic presentation.
Furthermore, tumors in the OPN cohort more often
were malignant, were larger on average, and more
were centrally located or involved a solitary kidney
(see Table 2). These observations suggest that in
this study patient selection and tumor biology
were substantially different in the two groups,
with all comparisons reflecting a higher-risk population for OPN.
Table 2
Comparison of patient characteristics, perioperative parameters, and outcome in OPN and LPN
No. patients
ECOG performance status R 1 (%)
Mean preoperative serum creatinine (mg/dL)
% Symptomatic at presentation
Mean tumor size (cm)
% central tumors
% solitary kidney
Mean operative time (min)a
Mean warm ischemia time (min)
Mean blood loss (cm3)a
% with RCC on final pathologya
Mean hospital stay (days)a
% intraoperative complications
% postoperative urologic complicationsa
% postoperative hemorrhagea
% postoperative urine leak
% patients requiring subsequent procedurea
OPN
LPN
1029
14.7
1.25
33.5
3.5
53.3
21.6
266
20.1
376
83
5.8
1
5
1.6
2.3
3.5
771
1.4
1.01
8.8
2.7
34.4
4.2
201
30.7
300
72
3.3
1.8
9.2
4.2
3.1
6.9
649
650
Nguyen et al
small renal masses proactively and seems to be
associated with reduced morbidity and improved
quality of life.4648 Established ablative techniques
include cryoablation and radiofrequency ablation
(RFA), both of which can be performed laparoscopically or percutaneously. The laparoscopic approach is preferred when mobilization away from
adjacent organs is required. The percutaneous approach is even less invasive and is particularly
suited to posteriorly located tumors.49
Because of the relatively recent application of
thermal ablation to renal tumors, the long-term oncologic efficacy of these techniques has not been
established to the same degree as surgical excision. There currently is greater experience with
cryoablation, and there now are some studies
with at least 5 years of follow-up data that suggest
durable cancer control. Hegarty and colleagues50
reported on 66 patients, all of whom had at least
5 years of follow-up after laparoscopic renal cryoablation, and demonstrated 5-year overall and
cancer-specific survival rates of 81% and 98%,
respectively. Another series of 48 patients with
minimum and median follow-ups of 3 years and
64 months, respectively, demonstrated a cancerspecific survival of 100%.51 RFA is less established than cryoablation, and its technology is still
evolving; a number of different RFA generators
and probes are commercially available.52 A review
of the literature indicates that RFA can be effective
in treating small renal masses, providing cancerspecific survival ranging from 83% to 100% at
a mean follow-up of 20 months.53 Long-term
data (R 5 years) are required to establish the
true oncologic efficacy of RFA and the optimal
method for delivering radiofrequency energy.
Because it is less invasive, thermal ablation has
gained considerable traction in the field recently.
This technology comes with several potential limitations that must be considered carefully during
patient counseling and clinical decision making,
however. The current literature suggests that local
cancer control with thermal ablation is inferior to
that achieved with PN. Recent meta-analyses reported higher local recurrence rates with cryoablation and RFA than with surgical excision: 4.6%,
7.9%, and 2.7%, respectively, in the study by
Weld and Landman54 and relative rates of 7.45,
18.23, and 1.0, respectively, in the study by Kunkle
and colleagues.55 Furthermore, the validity of the
radiographic definition of postablative success
has been called into question, with recent data
demonstrating that a small percentage of patients
are found to have viable cancer on biopsy of an
ablated tumor despite lack of enhancement on
MRI.56 Such data suggest that recurrence may
be more common after thermal ablation,
particularly after RFA, than previously appreciated. Another disadvantage of thermal ablation
is the lack of a pathologic diagnosis following
treatment and an inability to confirm complete
tumor kill.
Most local recurrences after thermal ablation
can be managed successfully with repeat ablation,
but indiscriminate use of thermal ablation in patients who otherwise may tolerate conventional
surgery may hinder subsequent salvage attempts.
A recent study from the Cleveland Clinic demonstrated that salvage surgery after previous ablation
can be challenging, and in many cases partial nephrectomy or laparoscopic surgery were not possible.57 A final limitation of thermal ablation relates
to tumor size: success rates fall substantially for
tumor diameters of 3.5 cm or greater. When all
these potential limitations are considered, thermal
ablation currently cannot be recommended as definitive therapy for the general patient population
and may be best suited for high-risk surgical candidates who have small, exophytic renal masses.
PN remains the preferred management for locally recurrent disease, but thermal ablation represents an attractive option for certain patients,
particularly those who have a history of previous
ipsilateral surgery, impaired renal function that
may be compromised further by hilar clamping,
or significant multifocal disease.68,69 Along these
lines, some have described a combined approach
using PN for dominant lesions and thermal ablation to manage remaining evident lesions, with
the goal of minimizing warm ischemic times while
rendering the kidney grossly free of disease.
651
652
Nguyen et al
option for NSS in select patients. It must be emphasized, however, that most tumors in a solitary
kidney can be managed safely and effectively
with PN, which is still the standard of care for
this patient population, assuming the patient is
a reasonable surgical candidate.
SUMMARY
The surgical treatment of localized RCC has undergone much change during the past decade.
Driven by the substantial evidence that preservation of renal function is a relevant clinical consideration for all patients, increasing efforts have been
made to employ nephron-sparing approaches
whenever possible. Efforts that once focused on
avoiding dialysis now should be directed toward
optimizing renal function. The prevailing data indicate that PN provides effective curative treatment
for localized renal tumors on par with RN and
should be the first-line treatment option for most
patients, given requisite surgeon expertise. OPN
is particularly suited to complex situations such
as tumor in a solitary kidney or a central or hilar tumor, because this approach is more versatile and
because the current database suggests that perioperative morbidity is lower than with LPN. Currently, the use of LPN for a given clinical scenario
will depend primarily on patient selection and individual surgeon experience, but its growing importance in the armamentarium of cancer operations
cannot be denied. Thermal ablation is a novel modality with great potential, particularly in high-risk
surgical candidates, but its role as either a means
of definitive therapy or as a supplement to current
surgical approaches remains to be determined.
REFERENCES
1. Lam JS, Shvarts O, Pantuck AJ. Changing concepts
in the surgical management of renal cell carcinoma.
Eur Urol 2004;45:692705.
2. Cadeddu JA, Ono Y, Clayman RV, et al. Laparoscopic nephrectomy for renal cell cancer: evaluation
of efficacy and safety: a multicenter experience.
Urology 1998;52:7737.
3. Chan DY, Cadeddu JA, Jarrett TW, et al. Laparoscopic radical nephrectomy: cancer control for renal
cell carcinoma. J Urol 2001;166:20959.
4. Gill IS, Meraney AM, Schweizer DK, et al. Laparoscopic radical nephrectomy in 100 patients: a single
center experience from the United States. Cancer
2001;92:184355.
5. Permpongkosol S, Chan DY, Link RE, et al. Laparoscopic radical nephrectomy: long-term outcomes.
J Endourol 2005;19:62833.
6. Russo P. Open partial nephrectomy: an essential
operation with an expanding role. Curr Opin Urol
2007;17:30915.
7. Lau WK, Blute ML, Weaver AL, et al. Matched comparison of radical nephrectomy vs nephron-sparing
surgery in patients with unilateral renal cell carcinoma and a normal contralateral kidney. Mayo Clin
Proc 2000;75:123642.
8. McKiernan J, Simmons R, Katz J, et al. Natural history of chronic renal insufficiency after partial and
radical nephrectomy. Urology 2002;59:81620.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
653
654
Nguyen et al
41. Zorn KC, Gong EM, Orvieto MA, et al. Comparison of
laparoscopic radical and partial nephrectomy: effects
on long-term serum creatinine. Urology 2007;69:
103540.
42. Link RE, Permpongkosol S, Gupta A, et al. Cost
analysis of open, laparoscopic, and percutaneous
treatment options for nephron-sparing surgery. J Endourol 2006;20:7829.
43. Gill IS, Colombo JR Jr, Moinzadeh A, et al. Laparoscopic partial nephrectomy in solitary kidney. J
Urol 2006;175:4548.
44. Gill IS, Colombo JR Jr, Frank I, et al. Laparoscopic partial nephrectomy for hilar tumors. J Urol 2005;174:
8503.
45. Frank I, Colombo JR Jr, Rubinstein M, et al. Laparoscopic partial nephrectomy for centrally located renal tumors. J Urol 2006;175:84952.
46. Hinshaw JL, Lee FT Jr. Image-guided ablation of renal cell carcinoma. Magn Reson Imaging Clin N Am
2004;12:42947.
47. Lowry PS, Nakada SY. Renal cryotherapy: 2003 clinical status. Curr Opin Urol 2003;13:1937.
48. Murphy DP, Gill IS. Energy-based renal tumor ablation: a review. Semin Urol Oncol 2001;19:13340.
49. Hafron J, Kaouk JH. Ablative techniques for the
management of kidney cancer. Nat Clin Pract Urol
2007;4:2619.
50. Hegarty NJ, Gill IS, Kaouk JH, et al. Renal cryoablation: 5 year outcomes. [abstract 1091]. J Urol 2006;
175(Suppl):351.
51. Davol PE, Fulmer BR, Rukstalis DB. Long-term
results of cryoablation for renal cancer and complex renal masses. Urology 2006;68(1 Suppl):
S26.
52. Mulier S, Miao Y, Mulier P, et al. Electrodes and multiple electrode systems for radiofrequency ablation:
a proposal for updated terminology. Eur Radiol
2005;15:798808.
53. Park S, Cadeddu JA. Outcomes of radiofrequency
ablation for kidney cancer. Cancer Control 2007;14:
20510.
54. Weld KJ, Landman J. Comparison of cryoablation,
radiofrequency ablation and high-intensity focused
ultrasound for treating small renal tumours. BJU Int
2005;96:12249.
55. Kunkle DA, Egleson BL, Uzzo RG. Excise, ablate, or
observe: the small renal mass dilemmaa metaanalysis and review. J Urol 2008;179:122733.
56. Hegarty NJ, Kaouk J, Remer EM, et al. Lack of enhancement on 6-month MRI does not guarantee
complete cancer cell kill following radiofrequency
ablation of small renal tumors. [abstract 1718].
J Urol 2006;175(4 Suppl):552.
57. Nguyen CT, Lane BR, Kaouk J, et al. Surgical
salvage of renal cell carcinoma recurrence
after thermal ablative therapy. J Urol 2008;180:
1049.
655
Resection of Renal
Tumors Invading
the Vena Cava
Chad Wotkowicz, MD*, Matthew F.Wszolek, MD,
John A. Libertino, MD
KEYWORDS
Renal cell carcinoma Inferior vena cava
Surgical management
CLINICAL PRESENTATION
The advent of cross-sectional imaging for the
work-up of abdominal pain has resulted in an
increase of RCC prevalence. A number of patients
who have venous tumor thrombus can be asymptomatic depending on the level of the tumor
thrombus and the extent of occlusion of the IVC.
Significant venous congestion as a result of caval
intrusion can present with varying symptoms,
including significant lower extremity edema, varicocele formation, proteinuria, caput medusae,
and even pulmonary emboli. If the tumor extends
above the level of the hepatic veins, Budd-Chiari
syndrome may result from obstruction of the major
hepatic veins, resulting in a triad of hepatomegaly,
abdominal fullness/pain, and ascites. The resulting
varices produce massive collaterals with associated impaired hepatic function and portal hypertension. Additional symptoms of RCC include
flank discomfort, hematuria, and constitutional
changes (fever, weight loss, fatigue). These constitutional symptoms usually indicate the presence of
metastatic disease with an overall poor prognosis.
urologic.theclinics.com
658
Wotkowicz et al
patients the authors preferred imaging modality is
MRI, specifically magnetic resonance venography,
in combination with CT studies and threedimensional reformatted images (Fig. 1). Tumor
at or above the level of the diaphragm requires
transesophageal echocardiography and may
necessitate angiography to delineate the extent
of the tumor thrombus. At the present time positron emission tomography (PET) scans have
a limited diagnostic role; however, this modality
is being evaluated at the Lahey Clinic as part of
the preoperative evaluation and postoperative
follow-up. The authors have noted that PET CT
has demonstrated lesions in the liver that have
not been detected on ordinary CT or MRI (Fig. 2).
A report from Zini and colleagues12 suggests
that preoperative measurements of the renal vein
and IVC diameter with associated tumor thrombus
correlate with the rate of renal ostial wall invasion
and may serve as another prognostic indicator.
The importance of preoperative imaging for surgical planning cannot be overemphasized. Tumor
thrombus extending to the level of the hepatic
veins or higher may require cardiopulmonary bypass and circulatory arrest to provide insurance
against excessive blood loss. Patients slated for
cardiopulmonary bypass and circulatory arrest
should have a cardiac evaluation, which may
include stress testing or a coronary angiogram. If
significant coronary artery disease is discovered,
it may be treated with either a stent or bypass
grafting. In five of the authors patients bypass
grafting was performed concomitantly with the
radical nephrectomy and IVC thrombectomy.13
RENAL ANGIOINFARCTION
Although only one prospective trial of preoperative
angioinfarction is available to validate its use, the
authors find that preoperative renal artery embolization is an important adjunctive tool in the treatment for advanced RCC, including patients who
have venous tumor involvement.15 Preoperative
renal angioinfarction facilitates the dissection of
the renal tumor as a result of local tissue edema
from hypoxia and tissue necrosis. In addition, it
potentially may decrease the extent of the tumor
thrombus while minimizing intraoperative blood
loss associated with extensive venous collaterals.
Renal angioinfarction also allows the surgeon to
ligate or transect the renal vein before controlling
or occluding the renal artery. Clinicians must be
aware of the postinfarction syndrome caused by
innate and humoral immune responses to the
infracted kidney.16 This syndrome is characterized
by chills, fevers, flank pain, malaise, hematuria,
transient hypertension, and hyponatremia, all of
which are self limiting.17
Fig. 1. (A) Coronal scan demonstrates the extent of tumor thrombus to the level of the major hepatic veins. (B)
Lateral reconstructions indicate thrombus above the diaphragm. Cardiopulmonary bypass was required to resect
this tumor.
Box 1
American Joint Committee on Cancer 2002 TNM
staging system for renal cell carcinoma
T: Primary tumor
Tx: Primary tumor cannot be assessed
T0: No evidence of primary tumor
T1: Tumor <7 cm in diameter, limited to kidney
T1a: Tumor 04 cm in greatest diameter, confined to kidney
T1b: Tumor 47 cm in greatest diameter, confined to kidney
T2: Tumor > 7 cm in greatest diameter, confined
to kidney
T3: Tumor extends into major veins or invades
adrenal gland or perinephric tissues but not beyond Gerotas fascia
T3a: Tumor directly invades adrenal gland or
perirenal and/or renal sinus fat but not beyond
Gerotas fascia
Fig. 2. PET scan obtained during preoperative evaluation for planned radical nephrectomy demonstrates
increased uptake in multiple sites of the liver.
659
Wotkowicz et al
660
60
50
40
30
20
10
0
t
0
0
0
5
5
5
5
-9
-8
en
-7
-8
-9
00
00
86
-2
76
es
71
81
91
-2
r
9
6
9
9
9
9
1
1
9
1
1
1
1
0
-p
19
20
06
20
661
662
Wotkowicz et al
remove the kidney and thrombus as one specimen
(Fig. 12). The advantage of bypass is an essentially
bloodless operative field, but the authors
recognize and accept the complications of bypass, including coagulopathy and the potential
for neurologic complications.
VENOVENOUS BYPASS
Patients who have minimal extension of thrombus
above the level of the diaphragm can be managed
with venovenous bypass via a caval-atrial
shunt.31,32 With this approach the vena cava
needs to be controlled at the infrarenal level, at
the level of both renal veins, and at its intrapericardial portion. Once control is established, the
cannulas can be placed in the right atrium or
axillary vein and the femoral veins, and bypass
can be initiated before cavotomy. Bleeding from
the hepatic venous system can be managed by
cross-clamping the hepatic veins or by the Pringle
maneuver. The Pringle maneuver can be used for
up to 45 minutes before liver metabolism is
affected significantly. Although this technique
avoids cardiopulmonary bypass and circulatory
arrest, the incidence of hepatic venous bleeding
can be significant.
663
664
Wotkowicz et al
described by the authors group and by Ciancio
and colleagues.3335 The liver is mobilized to the
left after the division of the ligamentum teres, falciform ligament, triangular ligament, and superior
coronary ligament of the liver. The porta hepatis
is accessed via the foramen of Winslow, and the
Pringle maneuver is employed. This technique
provides excellent access to the retrohepatic portion of the vena cava and allows mobilization of the
liver from the vena cava, leaving only the major
hepatic veins in continuity. After liver mobilization
the surgeon can palpate and milk the tumor thrombus caudally below the confluence of the hepatics
veins to limit hepatic venous congestion associated with hepatic clamping. All cases are performed with the use of transesophageal echo
monitoring.
Fig. 10. Right atriotomy exposing tumor thrombus after circulatory arrest has been achieved.
Fig. 11. (A, B) Schematic demonstrating the removal of atrial tumor thrombus. When tumor burden is too large,
the atrial component may be fractured and removed first.
ENDOLUMINAL OCCLUSION
AND CAVAL THROMBUS
665
666
Wotkowicz et al
treating tumors extending into the IVC.40 Urologists at the Cleveland Clinic successfully removed
a right atrial tumor thrombus in a calf model using
combined laparoscopy and thoracoscopy with
deep hypothermic circulatory arrest.41 Such
approaches are sure to surface with the continuing
rapid advances in minimally invasive surgical
techniques.
NEPHRON-SPARING SURGERY
AND TUMOR THROMBUS
The unfortunate patient who has a solitary kidney
and RCC with associated tumor thrombus and
decent performance status may be a candidate
for nephron-sparing surgery. Sengupta and colleagues4244 have reported their experience as
well as additional case reports. These studies
showed limited oncologic success and a relatively
high rate of eventual completion radical nephrectomy. The authors recommend that urologists
attempting these procedures be skilled in
extracorporeal bench surgery, renal autotransplantation, and vena caval reconstruction with
the caveat that achieving negative margins is
more important than avoiding the need for renal
replacement therapy.
bland tumor thrombus in the setting of RCC to prevent unwanted pulmonary embolism. The authors
reviewed 160 patients who had thrombus extending 2 cm and more above the renal vein and identified 40 patients who had total or partial venous
occlusion and the presence or absence of associated bland thrombus. (It should be noted that any
attempt to resect or ligate the IVC should be preceded by efforts to preserve the integrity of the
lumbar drainage system.) Blute and colleagues
recommend ligating no more than two lumbar
veins. Patients who have a patent cava and no
associated distal or bland tumor thrombus can
be managed with cavatomy closure only. A partially occluded vena cava with distal pelvic bland
thrombus can be managed with an interruption
caval filter. Patients who have a totally occluded
vena cava with associated bland thrombus are
treated by IVC staple ligation. In the latter group,
distal margins should be sent to pathology for frozen-section analysis. The outcomes for these
groups fail to demonstrate any significant morbidity and thus support the use of these techniques in
the management of retrograde bland tumor thrombus in this complex surgical population.
COMPLICATIONS
The incidence of complications often depends on
the level of tumor thrombus and the surgical
approach taken. Boorjian and colleagues47
reviewed their experience with more than 650
patients undergoing nephrectomy and tumor
thrombectomy and noted that the incidence of
early (<30 days) and late complications correlated
with thrombus level. Operative time and blood loss
followed the same trend. The present authors have
reported their experience with minimal-access
Table 1
Operative and perioperative comparison of traditional (TMS) versus minimal-access (MA) cardiopulmonary
bypass26
Operation
Cardiopulmonary
bypass time
Circulatory arrest
Days ventilated
Length of stay
Transfusions
Overall complications
Pulmonary
Cardiac
Renal
Infection
Hepatic
MA Median (n 5 28)
P-value
600 (295995)
135 (50217)
450 (270761)
148 (86265)
<.001
.527
33 (1290)
7 (1110)
26 (2114)
11 (450)
17
12
12
6
10
7
34 (1762)
4 (146)
12 (545)
5 (215)
21
7
13
4
7
5
.880
.032
.007
.002
1.000
.264
.741
.311
.210
.331
Data from Wotkowicz C, Libertino J, Sorcini A, et al. Management of renal cell carcinoma with vena cava and atrial thrombus: minimal access vs median sternotomy with circulatory arrest. BJU Int 2006;98(2):28997.
CYTOREDUCTIVE NEPHRECTOMY
AND METASTECTOMY
Patients who have metastatic RCC face a poor
prognosis, with a median survival of 8 months
and a 2-year survival rate of 10% to 20%. A combined analysis of the two sentinel trials elucidating
the benefits of cytoreductive nephrectomy (Southwest Oncology Group-8949 and European Organization for Research and Treatment of Cancer)
revealed a survival benefit of 13.6 months for
nephrectomy combined with interferon-alpha therapy versus 7.8 months for interferon therapy
alone.48 Retrospective data from the UCLA group
suggest a more substantial survival benefit when
interferon alpha is replaced with interleukin-2.49
The underlying mechanisms of improved survival
with cytoreductive nephrectomy before systemic
therapy are not fully understood. The reduction in
growth factors, angiogenesis promoters, and
inhibitory immunomodulators by primary tumor
resection may enhance the efficacy of systemic
immunotherapy compared with immunotherapy
without prior cytoreductive nephrectomy.
First described by Barney and Churchill50 in
1939, the resection of pulmonary metastasis
remains an effective treatment for select patients.
Studies have attempted to define patient populations that would benefit from metastectomy
through subgroup analysis. Favorable prognostic
factors in resecting isolated pulmonary metastasis
include preoperative performance status, completeness of resection, number of lesions (fewer
than six), extent of lymph node involvement, and
length of disease-free interval. Patients having
synchronous lesions have significantly worse outcomes.51,52 Properly selected patients may have
5-year survival rates exceeding 50%.51 At the
present time, metastectomy in nonpulmonary sites
such as the liver and brain is controversial and
should be considered investigational.
CLINICAL OUTCOMES
There has been a modest increase in the number
of IVC thrombectomies performed by urological
oncologists and in improved survival outcomes.
These results can be attributed to improved imaging modalities and surgical planning as well as to
technological advances in intraoperative anesthesia and postoperative intensive care management.
Table 2 summarizes recent reports from tertiary
centers with significant experience in surgical
management of RCC with tumor thrombus. The
debate concerning the prognostic significance of
tumor thrombus level with regards to the current
TNM staging system may be resolved best by
a multi-institutional meta-analysis.5358
667
668
TumorThrombus
Stratification
Complications
(%)
Operative
Mortality (%)
659
19701989: 3.8
19902006: 2.0
Fijusawa 2007
55
3.6
Kalatte and
Belldegrun
2007
321
Level 1IV
I: infrahepatic (22)
II: intrahepatic (20)
III: suprahepatic (10)
IV: into atrium (3)
Renal vein: 166
IVC: 137
Atrium: 18
All complications
Renal vein; 12
IVC: 28
Atrium: 11
Renal vein: 2
IVC: 7
Atrium: 11
Author,Year
No. Patients
Blute 2007
Survival Outcomes:
Cancer-Specific Survival (CSS)
and Disease-Specific
Survival (DSS) (%)
5-year CSS
Level 0: 49.1
Level I: 31.7
Level II: 26.3
Level III: 39.4
Level IV: 7
Level 0 versus I (P 5 .002)
Level IIV (P 5 .868)
pN0/pNx, pM0: 59
pN1/pN2,pM1: 6
Histologic features
With tumor necrosis: 26
Without tumor necrosis: 61
With sarcomatoid: 3
Without sarcomatoid: 47
Perinephric fat invasion
Present: 32
Absent: 56
CSS (all levels)
1 year: 74.5
3 years: 51.4
5 years: 30.3
DSS (2/5/10-year)
Renal vein (58/41/24)
IVC (48/30/25)
Atrium (45/22/0)
Thrombus level (P 5 .53)
Overall immunotherapy
response rate: 19
Metastatic disease
Median survival: 16 months
Wotkowicz et al
Table 2
Surgical outcomes for patients who had renal cell carcinoma and tumor thrombus
Major complications: 6
Minor complications: 16
74
Major complications: 28
243
Renal vein: 87
IVC: 126
Atrium: 30
49
142
Libertino (unpublished
data)
Nevus classification
Level I: renal vein
Level II: infrahepatic IVC
Level III: retrohepatic to
diaphragm
Level IV:
supradiaphragmatic
Renal vein: 118
Subdiaphragmatic IVC: 24
669
670
Wotkowicz et al
SUMMARY
The surgical resection of large renal tumors with
associated tumor and bland thrombus within the
IVC presents a challenge to the urological surgeon. Given the magnitude of many of these procedures, surgeons who have experience at
tertiary centers are most adept in their management. The authors clinical experience is one of
the largest to date, and they hope this article
serves as guide to physicians treating this unique
population. They also commend their colleagues
who have encouraged alternative techniques
with equivalent outcomes that adhere to the principles of urologic oncology.
13.
14.
15.
16.
REFERENCES
1. Jemal A, Siegel R, Ward E, et al. Cancer statistics.
CA Cancer J Clin 2007;57(1):4366.
2. Lipworth L, Tarone R, McLaughlin J. The epidemiology of renal cell carcinoma. J Urol 2006;176(6 Pt 1):
23538.
3. Chow W, Devesa S, Waren J. Rising incidence of
renal cell cancer in the United States. JAMA 1999;
281(17):162831.
4. Jayson M, Sanders H. Increased incidence of serendipitously discovered renal cell carcinoma. Urology
1998;51(2):2035.
5. Marshall F, Dietrick D, Baumgartner W, et al. Surgical
management of renal cell carcinoma with intracaval
neoplastic extension above the hepatic veins.
J Urol 1988;139(6):116672.
6. Skinner DG, Pfister RF, Colvin R. Extension of renal
cell carcinoma into the vena cava: the rationale for
aggressive surgical management. J Urol 1972;
107(5):7116.
7. Neves R, Zincke H. Surgical treatment of renal cancer with vena cava extension. Br J Urol 1987;59(5):
3905.
8. Montie J, el Amnar R, Pontes J, et al. Renal cell carcinoma with inferior vena cava tumor thrombi. Surg
Gynecol Obstet 1991;173(2):10715.
9. Swierzewski D, Swierzewski M, Libertino J. Radical
nephrectomy in patients with renal cell carcinoma
with venous, vena caval, and atrial extension. Am
J Surg 1994;168(2):2059.
10. Emmott R, Hayne L, Katz I, et al. Prognosis of renal
cell carcinoma with vena cava and renal vein
involvement: an update. Am J Surg 1987;154(1):
4953.
11. Hatcher P, Anderson E, Paulson D, et al. Surgical
management of renal cell carcinoma invading the
vena cava. J Urol 1991;145(1):203.
12. Zini L, Destrieux-Garnier L, Leroy X, et al. Renal vein
ostium wall invasion of renal cell carcinoma with an
inferior vena cava tumor thrombus: prediction by
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
57.
58.
671
Radical nephrectomy and regional lymphadenectomy have been the cornerstone of therapy for renal cell carcinoma (RCC) for several decades;1
however, debate regarding the potential advantages of lymph node dissection for RCC continues.
Currently, there are no definitive data indicating
a survival advantage to lymphadenectomy. Furthermore, systematic complete lymph node dissection adds time to the procedure and requires
manipulation of the great vessels, which some surgeons may find challenging. This article examines
the rationale for lymphadenectomy in the management of renal cell carcinoma and reviews the limited literature on the subject.
Carcinoma of the kidney and renal pelvis is expected to be newly diagnosed in over 54,000 patients in the United States and will result in over
13,000 deaths, accounting for approximately 3%
of all cancer deaths in 2008.2 About one-third of
new patients presenting with RCC have metastatic
disease. Another third of patients presenting with
localized disease eventually experience recurrence and progression. Approximately 25% of patients with metastatic RCC have clinically evident
lymphadenopathy. While metastatic disease is
highly resistant to chemotherapy, systemic therapy options now include targeted therapy in addition to immunotherapy.3 Thankfully, survival for
patients with RCC appears to be improving, with
decreasing death rates per 100,000 from 6.16 to
5.91 in men and from 2.95 to 2.72 in women between the early 1990s and today.2 This is reflected
in improved 5-year survival rates from 52% between 1974 and 1976 to 63% in 1999.4 Positive
nodes have been clearly shown to have an
independent adverse effect on outcome, regardless of other prognostic factors.57 For patients
with node-positive disease, 5-year survival rates
range between 5% and 35%. Most studies of
node-positive renal cell carcinoma report 5-year
survival rates of about 15%.8 Fig. 1 demonstrates
the impact of lymph node status on cancer-specific survival among patients treated surgically for
RCC at the Mayo Clinic.
Proponents of lymphadenectomy point to higher
survival rates for patients undergoing radical nephrectomy plus extended lymph node dissection,
compared with historical studies that did not include routine lymphadenectomy. Opponents point
to the high rates of hematogenous metastases and
question the value of lymph node dissection in a
disease that follows an unpredictable course. To
definitively address the potential benefit of lymphadenectomy in RCC, the European Organization for
Research and Treatment of Cancer Genitourinary
Group launched a head-to-head randomized
phase 3 trial at multiple European centers in 1988.
The trial, which completed enrollment in 1992, is
designed to compare the long-term results of radical nephrectomy with complete lymphadenectomy
(n 5 383) against radical nephrectomy alone (n 5
389) in patients without evidence of metastases.
Early results indicate that complete lymph node
dissection did not increase morbidity associated
with radical nephrectomy. Pathology confirmed
lymph node metastases in 3.3% of clinically negative nodes after lymphadenectomy. However, survival in the study so far had been reported to be
excellent overall and more follow-up time is needed
to compare tumor-free survival and overall survival
urologic.theclinics.com
674
Fig.1. Cancerspecific survival from time of radical nephrectomy by lymph node status.
between the lymphadenectomy and non-lymphadenectomy groups.9 While results are pending, debate will continue regarding the need for
lymphadenectomy in RCC patients.
Although the role of nephron-sparing surgery for
renal masses under 7 cm continues to evolve,10,11
the appropriate surgical treatment for large renal
masses has not changed substantively since Robson and colleagues1 first reported increased survival in a small cohort of patients who received
lymphadenectomy in 1969. Surgical excision of
solid tumors was common by the late nineteenth
century.12 After Halsted demonstrated the efficacy
of extensive regional lymphadenectomy for breast
cancer in 1894, radical excision and regional lymphadenectomy gradually evolved as the standard
of care for most carcinomas. During the first half
of the twentieth century simple nephrectomy became the standard treatment for localized RCC.
The procedure typically involved removal of the
kidney from the surrounding Gerotas fascia. Surgeons generally left the perirenal fat, adrenal
gland, and regional lymph nodes in situ.
The first radical nephrectomy, removing the kidney, adrenal gland, and the surrounding Gerotas
fascia, was reported by Mortensen in 1948.13 The
reported rationale for the extent of surgery was
the observation that pathology studies revealed
perirenal fat infiltration in 13% of renal tumors;
therefore, removal of the fat and the organs contained within was postulated to increase survival.
In the 1960s, Robson and colleagues1,14 added retroperitoneal lymphadenectomy to radical nephrectomy and reported improved 5-year survival rates.
The first systematic survey of lymphatic drainage from the kidneys was published in 1935,
when Parker15 reported on the dissection of
cadavers and stillborn infants to detail the renal
lymphatic system. For the right kidney, the firstechelon nodes are the precaval, retrocaval, and
675
676
REFERENCES
1. Robson CJ, Churchill BM, Anderson W. The results
of radical nephrectomy for renal cell carcinoma.
J Urol 1969;101:297301.
2. Jemal A, Siegel R, Ward E, et al. Cancer statistics,
2008. CA Cancer J Clin 2008;58:7196.
3. Garcia JA, Rini BI. Recent progress in the management of advanced renal cell carcinoma. CA Cancer
J Clin 2007;57:11225.
4. Pantuck AJ, Zisman A, Dorey F, et al. Renal cell carcinoma with retroperitoneal lymph nodes. Impact on
survival and benefits of immunotherapy. Cancer
2003;97:29953002.
5. Frank I, Blute ML, Cheville JC, et al. An outcome prediction model for patients with clear cell renal cell
carcinoma treated with radical nephrectomy based
on tumor stage, size, grade and necrosis: the SSIGN
score. J Urol 2002;168:2395400.
6. Leibovich BC, Blute ML, Cheville JC, et al. Prediction
of progression after radical nephrectomy for patients
with clear cell renal cell carcinoma: a stratification
tool for prospective clinical trials. Cancer 2003;97:
166371.
7. Leibovich BC, Cheville JC, Lohse CM, et al. A scoring algorithm to predict survival for patients with
metastatic clear cell renal cell carcinoma:
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
677
678
Surgical Intervention
in Patients with
Metastatic Renal Cancer :
Metastasectomy
and Cy toreductive
Nephrectomy
Paul Russo, MD, FACSa,*, Matthew Francis OBrien, MDb
KEYWORDS
Renal cancer Cytoreductive
Kidney cancer Metastasectomy
a
Department of Surgery, Urology Service, Weill Cornell College of Medicine, Memorial Sloan Kettering Cancer
Center, 1275 York Avenue, New York, N Y 10021, USA
b
Urological Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, N Y 10021, USA
* Corresponding author.
E-mail address: russop@mskcc.org (P. Russo).
urologic.theclinics.com
There will be an estimated 54,390 new renal tumors and 13,010 deaths from renal cancer in
the United States in 2008.1 Approximately 30%
to 40% of patients with malignant renal cortical
tumors will either present with or later develop
metastatic disease. In metastatic renal cancer,
surgical intervention may be performed alone or
in combination with systemic therapy. Although
the evolution of metastatic disease usually occurs
within 2 years after the radical or partial nephrectomy, disease-free intervals of up to 30 years and
metastases to unusual sites (eg, endocrine
glands, digits) can cause diagnostic dilemmas.
Unlike patients with renal cortical tumors detected incidentally during abdominal imaging obtained for other reasons, the vast majority of
patients with metastatic renal tumors have large,
locally advanced tumors often with regional nodal
and or renal vein or inferior venal extension. Approximately 90% of the metastatic renal tumors
are of the conventional clear cell histologic subtype. Unfortunately for metastatic renal cancer
patients, systemic chemotherapy, cytokine therapy, and hormonal manipulations alone or in
combination have low overall response rates
(10%15%) and are rarely associated with
680
METASTASECTOMY
In 1939 Barney and Churchill5 first reported a patient who underwent nephrectomy and a resection
of an isolated pulmonary metastasis for a renal
cancer only to die 23 years later of coronary artery
disease. Over the last 60 years, the surgical resection of limited metastatic disease (metastasectomy) was offered to patients and selectively
performed in the absence of effective systemic
therapies. The reported selection criteria for this
aggressive surgical approach varied from study
to study and reported significant prognostic factors included the site and number of metastatic
deposits, completeness of resection, patient performance status, and the disease-free interval
from treatment of the primary tumor to the diagnosis of metastatic disease. Complete resection of
isolated metastases was associated with 5-year
survival rates of between 35% and 60%. Despite
successful resection of metastatic disease and
associated patient survival, these studies lacked
definitive proof that the surgical intervention itself,
as opposed to patient selection factors and the
natural history of renal cancer, led to the observed
outcomes.69 Pogrebniak and colleagues10 reported 23 patients who underwent resection of
pulmonary metastases from renal cell carcinoma
(RCC), 15 of whom had previously been treated
with IL-2based immunotherapy. Patients with resectable lesions had a longer survival (mean 49
months) than those patients with unresectable lesions (mean 16 months). Furthermore, in this
study, survival was not dependent upon the number of nodules removed. The investigators concluded that patients with metastatic RCC should
be offered an operation if the likelihood that complete resection of all sites of disease were high. Favorable subgroups include those patients with
a solitary site of metastases and disease-free interval to the development of metastases of greater
than 1 year. It should be noted that occasionally
sites of disease presumed to be metastatic RCC
are instead secondary tumors (eg, pancreatic islet
cell tumor) of either benign or malignant histology.
This diagnostic dilemma may be addressed in the
future with the further development of conventional clear cell specific immunopositron emission tomographic scanning with 124-I cG250
scanning.11
In a report from Memorial Sloan Kettering Cancer Center (MSKCC), prognostic factors associated with enhanced survival in 278 patients who
underwent surgical metastasectomy included
CYTOREDUCTIVE NEPHRECTOMY
The role of radical nephrectomy in patients with
extensive metastatic renal cancer, when complete
metastasectomy is not possible, has long been
debated. Given the lack of effective systemic therapies and the unpredictable natural history of metastatic RCC, many oncologists referred patients to
surgeons for resection of the primary tumor before
cytokine-based therapy. In theory, cytoreductive
radical nephrectomy is performed to remove
a large, potentially immunosuppressive, tumor
burden; to obtain accurate tumor histologic subtyping; and to prevent complications related to
the primary tumor during systemic therapy. On
rare occasions, a highly symptomatic tumor is removed for symptom palliation after the failure of
conservative palliation measures (eg, unremitting
gross hematuria or flank pain not relieved by
conservative care or angioinfarction). Radical nephrectomy should not be done to induce spontaneous remission, a phenomena observed only in
4 of 474 patients (0.8%) treated with radical nephrectomy alone in a study from the Cleveland
Clinic.16 Surgical mortality has been reported
from 2% to 11% for patients with large primary
renal tumors and metastatic disease. The possibility that the patient may not recover sufficiently to
receive systemic immunotherapy after preparatory
radical nephrectomy is of concern to surgeons and
medical oncologists alike. In a study of 195 patients with metastatic RCC treated at the National
Cancer Institute, 121 patients (62%) were eligible
for high-dose IL-2 following cytoreductive nephrectomy, leading to a response rate of 18%.
Yet, 38% of the patients in this series who underwent nephrectomy never received any immunotherapy either because of complications of
nephrectomy or because of rapid clinical deterioration from disease progression.17 Some oncology
groups recommend adjuvant radical nephrectomy
only if initial systemic therapy was effective in initiating clinical regression of metastatic sites. This
avoids the surgical morbidity.18,19
Two randomized and prospective clinical trials
have attempted to further define the role of cytoreductive nephrectomy in the treatment of metastatic renal cancerone in the United States
organized by the Southwest Oncology Group
(SWOG), and the other in Europe organized by
the European Organization Research and Treatment of Cancer (EORTC). Both used similar entry
criteria comparing treatment for metastatic renal
cancer with cytoreductive nephrectomy plus interferon alpha-2b versus interferon alpha-2b alone.
In the SWOG trial, between 1991 and 1998, 246
patients with metastatic renal cancer and with the
tumor-bearing kidney in place from 80 participating institutions were randomly assigned to the
two groups of 123 patients each. Eligible patients
had a histologically confirmed diagnosis of metastatic RCC in tissue obtained by needle biopsy
or aspiration of a least one measurable metastatic
681
682
Percent survival
low-risk (0)
intermediate (1-2)
high (3-5)
80
60
40
20
0
20
40
60
80
100
683
684
Fig. 3. (A) Massive 21.2-cm left RCC in 43-year-old male with numerous pulmonary metastases started on sunitinib,
50 mg/d. (B) Ten-months later, near complete resolution in pulmonary metastases and marked reduction in left
kidney primary to 15 cm. Radical nephrectomy revealed viable poorly differentiated clear cell carcinoma with
areas of necrosis.
conventional clear cell cancers.31,32 For the papillary and chromophobe carcinomas of the kidney,
patterns and sites of metastases are similar to
those for the conventional clear cell carcinoma,
but the molecular mechanisms are likely different.33 For unknown reasons, metastastic papillary
and chromophobe carcinomas are virtually unresponsive to systemic cytokine and TKI therapy.34
A complete description of these clinical trials is
available at ClinicalTrials.gov.
Medical oncologists and surgeons alike have
noticed remarkable regression of both the metastatic sites and primary tumor following TKI (sunitinib and sorafenib) administration and, in some
cases, resolution of distant metastases followed
by marked regression of the primary tumor, allowing for radical nephrectomy to be integrated into
the patient management of patients felt to be previously unresectable (Fig. 3). However, during the
break from TKI administration during the period
of postnephrectomy recovery, rapidly recurrent
distant metastatic disease has been noticed. Reinitiation of the TKI has lead to secondary response
in the same metastatic sites, suggesting that in
these patients chronic administration of TKI is required. In a recent report from MD Anderson Cancer Center, 44 patients treated with multitargeted
therapies before cytoreductive nephrectomy and
resections of locally recurrent RCC were compared with 58 matched patients treated with cytoreductive nephrectomy or resection alone. In this
study, 27.5% of the nephrectomies were done
using laparoscopic techniques. Complications
occurred in 39% of patients treated with preoperative TKI versus 28% treated with operation alone
(P 5 .287). The investigators concluded that the
preoperative treatment of patients before cytoreductive nephrectomy and resection does not significantly increase operative morbidity, which
was substantial in both groups, and, although the
ultimate value of this approach remains to be determined, it appears to be a comparable to cytoreductive nephrectomy and resection alone.35
SUMMARY
RCC represents a family of neoplasms possessing
unique molecular and cytogenetic defects with
90% of the metastases emanating from the conventional clear cell carcinoma subtype. For patients with metastatic renal cancer, prognostic
factors defined in systemic therapy clinical trials
stratify patients into good, intermediate, and
poor risk groups with median survival varying
from 4 to 13 months. These same factors also
stratify patients whose renal cancers were initially
resected completely and who then developed
subsequent metastatic disease. Metastasectomy
performed in low-risk patients was significantly associated with enhanced survival when compared
with low-risk patients not undergoing metastasectomy. Careful case selection by surgeons of relatively healthy patients with disease-free intervals
of greater than 1 year, which is associated with
subsequent survival, makes it difficult to distinguish between the natural history of metastatic renal cancer and direct therapeutic effect. Two
randomized, prospective clinical trials from the
EORTC and SWOG, analyzed both separately
and then together, demonstrated a modest survival advantage of approximately 6 months for patients undergoing cytoreductive nephrectomy
followed by interferon alfa-2b. The mechanism
by which the cytoreductive nephrectomy benefits
the patient is not known but may relate to removing an immunosuppressive tumor burden. It is
our opinion that, once effective systemic agents
are developed, both metastasectomy and cytoreductive nephrectomy will play greater roles in consolidating clinical responses. Numerous adjuvant
REFERENCES
1. Jemal A, Siegel R, Ward E, et al. Cancer statistics,
2008. CA Cancer J Clin 2008;58:7196.
2. Motzer RJ, Russo P. Systemic therapy for renal cell
carcinoma. J Urol 2000;163:40817.
3. Bukowski RM, Wood LS. Renal cell carcinoma: state
of the art diagnosis and treatment. Clin Oncol 2008;
11:921.
4. OBrien F, Motzer R, Russo P. Sunitinib therapy in renal cell carcinoma. BJU Int 2008;101:133942.
5. Barney JD, Churchill EJ. Adenocarcinoma of the kidney with metastasis to the lung: cured by nephrectomy and lobectomy. J Urol 1939;42:26976.
6. Giuliani L, Giberti C, Martorana G, et al. Radical extensive surgery for renal cell carcinoma: long-term results and prognostic factors. J Urol 1990;143:46873.
7. Golimbu M, Joshi P, Sperber A, et al. Renal cell carcinoma: survival and prognostic factors. Urology
1986;27:291301.
8. Maldazys JD, deKernion JB. Prognostic factors in
metastatic renal carcinoma. J Urol 1986;136:3769.
9. Neves RJ, Zincke H, Taylor WF. Metastatic renal cell
cancer and radical nephrectomy: identification of
prognostic factors and patient survival. J Urol
1988;139:11736.
10. Pogrebniak HW, Haas G, Linehan WM, et al. Renal
cell carcinoma: resection of solitary and multiple metastases. Ann Thorac Surg 1992;54:338.
11. Divgi CR, Pandit-Taskar N, Jungbluth AA, et al. Preoperative characterization of clear-cell carcinoma
using iodine-124-labelled antibody chimeric G250
(124I-cG250) and PET in patients with renal masses:
a phase I trial. Lancet Oncol 2007;8:30410.
12. Kavolius JP, Mastorakos DP, Pavolvich C, et al. Resection of metastatic renal cell carcinoma. J Clin
Oncol 1998;16:22616.
13. Wronski M, Russo P, Galicich J, et al. Surgical resection of brain metastases from renal cell carcinoma.
Urology 1996;47:18793.
14. Kim SH, Brennan MF, Russo P. The role of surgery in
the treatment of adrenal metastasis. Cancer 1998;
82:38994.
15. Russo P, Snyder M, Vickers A, et al. Cytoreductive
neprhectomy and nephrectomy/complete metastasectomy for metastatic renal cancer. The Scientific
World Urology 2007;2:4252.
16. Montie JE, Stewart BH, Straffon RA, et al. The role of
adjuvant nephrectomy in patients with metastatic
renal cell carcinoma. J Urol 1977;117:2725.
17. Walther MM, Yang JC, Pass HI, et al. Cytoreductive
surgery before high dose interleukin-2 based therapy in patients with metastatic renal cell carcinoma.
J Urol 1997;158:16758.
18. Rackley R, Novick A, Klein E, et al. The impact of
adjuvant nephrectomy on multimodality treatment
of metastatic renal cell carcinoma. J Urol 1994;
152:1399403.
19. Sella A, Swanson DA, Ro JY, et al. Surgery following
response to interferon-alpha-based therapy for
residual renal cell carcinoma. J Urol 1993;149:1921.
20. Flanigan RC, Salmon SE, Blumenstein BA, et al. Nephrectomy followed by interferon alfa 2-b compared
with interferon alfa-2b alone for metastatic renal cell
carcinoma. N Engl J Med 2001;345:16559.
21. Mickisch GH, von Poppel H, de Prijck L, et al. Radical nephrectomy plus interferonalfa based immunotherapy compared with interferon alfa alone in
metastatic renal-cell carcinoma: a randomized trial.
Lancet 2001;358:96670.
22. Flanigan RC, Mickisch G, Sylvester R, et al. Cytoreductive npherectomy in patients with metastatic renal cancer: a combined analysis. J Urol 2004;171:
10716.
23. Bromwich E, Hendry D, Aitchison M. Cytoreductive
nephrectomy: Is it a realistic option in patients with
renal cancer. BJU Int 2002;89:5235.
24. Fossa SD. Interferon in metastatic renal cell carcinoma. Semin Oncol 2000;27:18793.
25. Motzer RJ, Mazumdar M, Bacik J, et al. Survival and
prognostic stratification of 670 patients with
advanced renal cell carcinoma. J Clin Oncol 1999;
17:285967.
26. Motzer RJ, Bacik J, Schwartz LH, et al. Prognostic
factors for survival in previously treated patients
with metastatic renal cell carcinoma. J Clin Oncol
2004;22:45463.
27. Kattan MW, Reuter VE, Motzer RJ, et al. A postoperative prognostic nomogram for renal cell carcinoma.
J Urol 2001;166:637.
28. Lee CT, Katz J, Fearn PA, et al. Mode of presentation
of renal cell carcinoma provides prognostic information. Urol Oncol 2002;7:13540.
29. Eggener SE, Yossepowitch O, Pettus JA, et al. Renal
cell carcinoma recurrence after nephrectomy for
localized disease. Predicting survival from time of
recurrence. J Clin Oncol 2006;24:310131.
30. Eggener SE, Yossepowitch O, Kudhu S, et al.
Objective (risk stratification) and subjective
(metastasectomy) factors impact prognosis in
patients with recurrent renal cell carcinoma following
nephrectomy. J Urol 2008;180(3):8738.
31. Iliopoulous O, Levy AP, Jiang C, et al. Negative
regulation of hypoxia-inducible genes by the von
685
686
34. Motzer RJ, Bacik J, Mariani T, et al. Treatment outcome and survival associated with metastatic renal
cell carcinoma of nonclear cell histology. J Clin
Oncol 2002;20:237681.
35. Margulis V, Matin SF, Tannir N, et al. Surgical
morbidity associated with administration of targeted
molecular therapies before cytoreductive nephrectomy or resection of locally recurrent renal cell carcinoma. J Urol 2008;180:948.
Systemic Therapy
for Metastatic Renal
Cell Carcinoma
Glenn S. Kroog, MD*, Robert J. Motzer, MD
KEYWORDS
Genitourinary Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial
Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
* Corresponding author.
E-mail address: kroogg@mskcc.org (G.S. Kroog).
Urol Clin N Am 35 (2008) 687701
doi:10.1016/j.ucl.2008.07.007
0094-0143/08/$ see front matter 2008 Elsevier Inc. All rights reserved.
urologic.theclinics.com
688
CYTOTOXIC CHEMOTHERAPY
The improved survival and quality of life for patients with metastatic RCC over the last several
years are direct results of advances in understanding the development of RCC. The genetics
of RCC tumorigenesis has been reviewed elsewhere68 and is briefly summarized here with attention to the consequences for systemic therapy
for RCC.
The von Hippel-Lindau (VHL) syndrome is an
autosomal dominant disease characterized by
the development of tumors in the cerebellum,
spine, retina, inner ear, pancreas, adrenal glands,
and kidneys. The kidney cancer in VHL syndrome
is uniformly clear cell RCC, and affected individuals have hundreds of clear cell RCCs per kidney.
The VHL tumor suppressor gene was identified in
1993. Both sporadic and inherited forms of clear
cell RCC are strongly associated with mutations,
deletions, or hypermethylations in the VHL gene
(VHL), which inactivate the gene. The VHL protein
functions as part of a multiprotein complex involved in targeting proteins for degradation by
marking them with ubiquitin. Major targets that
the VHL complex ubiquitinates are the transcription factors hypoxia-inducible factor 1a (HIF-1a)
and hypoxia-inducible factor 2a (HIF-2a).
Under normal oxygen conditions and with normal
VHL function, HIFs are degraded. When hypoxia
develops or if VHL is inactivated, HIF levels increase and HIF-dependent genes are transcribed
(Fig. 1). This leads to changes in expression of
various proteins and constitutes the cellular response to hypoxia. HIF levels can also be regulated
by growth factor and cell adhesion pathways,
leading to activation of the Ras-Rafmitogenactivated protein kinase pathway and the phosphatidylinositol 3-kinase-AKT-mTOR pathway.
The HIF-dependent response is characterized by
increased levels of VEGF, epidermal growth factor
receptor (EGFR), platelet-derived growth factor
(PDGF), glucose transporters (eg, GLUT-1), transforming growth factor-a (TGF-a, ligand for EGFR),
and erythropoietin. In the context of a clear cell
RCC, this results in stimulation of angiogenesis
and tumor cell proliferation. Because VEGF has
a central role during pathologic angiogenesis and
restricted expression in healthy adults, a variety
of therapeutic strategies aimed at blocking
VEGF-induced signal transduction have been
attempted. See Ferrara and colleagues9 for an excellent review of VEGF biology. Other RCC histologies are also associated with specific mutations.
For example, type 1 papillary RCC is characterized
by dysregulation or mutation in the tyrosine kinase
domain of the c-Met oncogene.
IMMUNOTHERAPY
Relapse of RCC many years after nephrectomy,
prolonged disease stabilization without systemic
treatment, and occasional spontaneous regressions suggested that host immune mechanisms
might control tumor growth. This led to the study
of immunotherapy for RCC.13 Interferon and IL-2
were reported to have antitumor activity in the
1980s and were the only proven therapy for metastatic RCC until recently. Immunotherapy for RCC
has been the subject of a recently updated analysis by Coppin and colleagues19 for the Cochrane
Collaboration. Combined data for a variety of
immunotherapies gave an overall response rate
of 12.4% (8.9% partial response and 3.5% complete response) compared with 2.4% in nonimmunotherapy control arms. High-dose IL-2 is
associated with increased vascular permeability
and requires inpatient monitoring, often in an intensive care unit. A limited number of centers
now offer high-dose IL-2. It has been associated
with a 4% incidence of treatment-related death
and should be offered only to patients with normal
B
angiogenesis
sunitinib
RTKs
RTKs
bevacizumab
VEGF
PDGF
Growth factors
nutrients
VEGF
PDGF
nutrients
RTKs
proliferation
HIF
sunitinib
Cancer cells
temsirolimus HIF
VEGF
PDGF
mTOR
VHL
RTKs
VEGF
PDGF
mTOR
HIF
VHL
HIF-responsive genes
HIF
HIF-responsive genes
VHL
HIF
Fig. 1. The VHL pathway in clear cell cancer of the kidney and inhibition by targeted therapies. Under normoxic
conditions, VHL promotes HIF degradation (box). When VHL mutates (A), HIF-dependent genes, including VEGF
and PDGF, are transcribed and angiogenesis is stimulated. Targeted therapies (B) block various aspects of these
events, leading to antiproliferative and antiangiogenic effects. Sunitinib and temsirolimus are shown as examples
of TKIs and mTOR inhibitors respectively. See text for details. RTKs, receptor tyrosine kinases.
689
690
TARGETED THERAPIES
VEGF antibodies, broad spectrum TKIs, and mTOR
inhibitors are generally referred to as targeted
therapies. However, most of the clinically useful
TKIs would be better described as multitargeted.
They are small molecules that bind to the ATP binding pocket of a group of evolutionarily related
kinases with varying affinities (Table 1). Some,
such as sorafenib (which also inhibits raf), have
an even broader spectrum of inhibition. The reason
or reasons for the varying efficacy in the clinic with
these agents is not clear. Therefore, although usually rationally designed, their use is therapy for RCC
is somewhat empiric. Targeted therapies have recently been reviewed.2830 Because the treatment
of RCC is in rapid flux, we focus on targeted therapies in detail. Results of major randomized phase III
trials are summarized in Table 2.
Sunitinib (SU11248)
Sunitinib was developed as an oral inhibitor of
VEGF receptor 2 and PDGF receptor b31 although
its spectrum of activity is broad. Sunitinib is primarily metabolized by cytochrome CYP3A4, resulting in formation of a major, pharmacologically
Table 1
Concentration required for 50% inhibition in vitro (IC50) for selected tyrosine kinase inhibitors
Kinase
VEGF receptor 1
VEGF receptor 2
VEGF receptor 3
PDGF receptor a
PDGF receptor b
EGFR
Fibroblast growth factor
receptor 1
Flt-3 and mutants
Stem cell factor receptor
(c-kit)
Ret
Colony stimulating factor 1
receptor/c-fms
Sunitinib
(nmol/L)31,90
Sorafenib
(nmol/L)91
Pazopanib
(nmol/L)92
Vatalanib
(PTK787/ZK 222,584)
(nmol/L)93
980
28
> 20,000
8302900
26
1590
20
57
>10,000
580
10
30
47
71
84
> 20,000
140
77
37
660
580
10300
110
3358
68
> 20,000
74
730
47
2800
146
1400
10.2 vs 5.4
649
Bevacizumab 1 interferon
vs interferon45
Setting
1st line
Good/intermediate
Good/intermediate
Good/intermediate
Intermediate/poor
10.9 vs
8.4 vs
7.3
5.5 vs 2.8
11 vs 5
5.5 vs 4.7 vs
3.1
80 vs 55
79 vs 55
32.1 vs
28.1 vs
15.5
77 vs 63
2 vs 0
31 vs 6
8.6 vs
8.1 vs
4.8
31 vs 13
903
750
626
Sorafenib vs placebo38
Sunitinib vs Interferon5
Temsirolimus vs temsirolimus
1 interferon vs interferon55
MSKCC Prognostic
Groups
Overall
Survival (mo)
Median Progression-Free
Survival (mo)
Dz Control
Rate (%)b
Overall Response
Rate (%)a
Number of
Patients
Drug(s)
Table 2
Major positive randomized phase III trials of targeted therapies in metastatic renal cell carcinoma
2nd line
1st line
1st line
691
692
Bevacizumab
Bevacizumab is a humanized monoclonal antibody
agent that binds and neutralizes all the major isoforms of VEGF-A.42 Bevacizumab was initially
investigated in a randomized, double-blind,
placebo-controlled phase 2 trial in patients with
clear cell carcinoma of the kidney.43 Bevacizumab
at doses of 3 and 10 mg per kilogram of body
weight or placebo was given every 2 weeks and
crossover from placebo to antibody treatment
was allowed. All patients had either received previous therapy with IL-2 or had a contraindication to
use of IL-2 and almost all had prior nephrectomy.
The primary end points were time to disease progression and response rate. A total of 116 patients
were randomly assigned to the three treatment
groups. At the time of a planned interim analysis,
the median time to progression was significantly increased to 4.8 months in the patients receiving the
10-mg/kg dose of bevacizumab, compared with
2.5 months for placebo. Four partial responses occurred, all in the group treated with bevacizumab at
10 mg/kg, for a partial response rate of 10%. Based
on the significant increase in time to progression,
accrual to the trial was stopped. The lack of an effect on survival was attributed to the crossover design. Most patients had VEGF levels below the
lower limit of detection and there was no significant
association between response and VEGF level.
However, the low sensitivity did not rule out a correlation between VEGF and clinical benefit.
Two randomized phase III studies examined
combinations of bevacizumab with interferon in
patients with predominantly clear cell histology.
A study comparing interferon alpha-2b (IFN-a-2b)
with or without bevacizumab coordinated through
the Cancer and Leukemia Group B (CALGB90,206; NCT00072046) has closed to accrual.44
Results of this study are not currently available.
The results of the Avoren trial was recently published and compared bevacizumab (10 mg/kg
every 2 weeks) plus IFN-a-2a (9 MU subcutaneously three times weekly; n 5 327) to placebo plus
IFN-a-2a (control group, n 5 322) in patients with
previously untreated metastatic RCC who had prior
nephrectomy.45 The study did not include a bevacizumab monotherapy arm because, at the time of
trial design, this was considered unethical. The
planned primary end point was overall survival.
Secondary end points included progression-free
survival and safety. An interim analysis of overall
survival was prespecified after 250 deaths. However, while the trial was in progress, new therapies
became available that could have confounded analyses of overall survival data, so the investigators
agreed with regulatory agencies that the
693
694
COMBINATION THERAPY
The available targeted and multitargeted therapies
represent a significant advance in the treatment of
RCC. However, none are associated with significant rates of long-term disease-free survival. Intensive research has begun over the last several
years to determine if combinations of targeted
therapies or targeted therapies plus immunomodulatory therapies are superior to sequential therapy with these agents.
Attempts to combine sunitinib with other agents
have been met with significant toxicity. A trial of
temsirolimus plus sunitinib was stopped due to
dose-limiting toxicity observed in two of three patients in the first cohort of a dose-finding study using temsirolimus 15 mg weekly and sunitinib 25 mg
daily.60 A phase I trial of sunitinib plus bevacizumab
has been performed. Hypertension was reported
to be the most common adverse event and grade
4 hemorrhage was observed in 2 out of 20 patients.61 Finally, a dose-finding study has been
completed combining sunitinib with interferon.62
The study enrolled 25 patients. Sunitinib 37.5 mg
per day (4 weeks on, 2 weeks off) and interferon 3
MU three times weekly was tolerated, although all
patients on the study experienced grade 3 adverse
events. Of the 6 patients at sunitinib 37.5 mg per
day and interferon 3 MU three times weekly, grade
3 adverse events included neutropenia, dyspnea,
hypertension, swelling face, and syncope. One patient at this dose level experienced a serious adverse event. For the entire group, 12% had partial
response and 80% stable disease. The investigators concluded that the toxicity of this regimen
does not justify continued study.
Sorafenib-based combinations have recently
been reviewed.63 Two phase II trials of sorafenib
plus interferon were published in 2007. Both used
sorafenib 400 mg twice daily plus IFN-a-2b 10
MU subcutaneously three times weekly and studied patients with similar prognosis to those treated
by Motzer and colleagues in the randomized study
of sunitinib versus interferon. Ryan and colleagues64 examined patients who had not received
prior systemic therapy and found responses in 12 of
62 patients (19%) and stable disease in 31 of 62 patients (50%). Median progression-free survival was
7 months. Toxicity was more severe than with either
agent alone. Fatigue (29%) and diarrhea (16%)
were the most common grade 3 toxicities. In
a smaller trial, Gollob and colleagues65 found
a 33% overall response rate (5% complete response and 28% partial response) and 45% stable
695
696
SEQUENCING OF AGENTS
Sunitinib/Sorafenib Sequencing
In two small series examining sequencing of sunitinib and sorafenib, some patients who progressed
Bevacizumab/Sorafenib or Sunitinib
In an expanded access program for sorafenib, the
subset of patients who had received prior bevacizumab was compared with the entire population.73
Partial response rate (3%), stable disease rate
(78%), and toxicity were similar to those for the entire population. Therefore prior bevacizumab treatment does not make patients less likely to have
stable disease in response to sorafenib. In addition, responses are seen to sunitinib after progression on bevacizumab. A phase II trial of 61 patients
reported 23% partial response and 57% stable
disease in patients treated with sunitinib who had
prior bevacizumab.74
Sorafenib/Bevacizumab or Tyrosine
Kinase Inhibitor
Similarly, small series suggest that prior sorafenib
does not preclude partial response or stable disease in patients subsequently treated with bevacizumab, sunitinib, or axitinib.75,76
of patients and the trial design, most trials evaluating currently approved targeted therapies have
been limited to or heavily biased toward clear
cell RCC. Even the phase III study of temsirolimus
included very few patients with documented non
clear cell histology. Experience is just being
obtained in nonclear cell histologies. For example, a recently published data set89 reported
patients with papillary or chromophobe RCC
treated with sunitinib or sorafenib as part of extended access programs at five cancer centers.
Out of 41 patients with papillary RCC, 13 received
sunitinib and 28 sorafenib. Two partial responses
were seen, both in the sunitinib group, and progression-free survival was 11.9 months in sunitinib-treated patients versus 5.1 months in
sorafenib-treated patients (P < .001). Out of 12
chromophobe RCC patients, 7 were treated with
sunitinib, and 5 with sorafenib. Two patients
treated with sorafenib and 1 patient treated with
sunitinib achieved a partial response, and stable
disease was achieved in the other patients. Sorafenib-treated patients tended to have a prolonged
median progression-free survival time (27.5
months). These data are preliminary and suggest
subtype-specific responses to different agents.
However, they are limited by small numbers of
patients and the absence of randomization. Further studies are ongoing.
SUMMARY
Until recently, only interferon and IL-2 were of
proven efficacy in the treatment of metastatic
RCC. Improved understanding of the biology of
RCC led to the development, study, and approval
by the FDA of sunitinib, sorafenib, and temsirolimus for treatment of metastatic RCC. Both sunitinib and temsirolimus are proven to be superior to
interferon. In addition, the combination of bevacizumab and interferon has also shown superiority
to interferon alone. However, none of the available
targeted therapies are associated with significant
rates of long-term disease-free survival. Clinical
research is ongoing to answer many questions
including:
What is the optimal sequence of available
agents?
Do combinations of targeted therapies provide additional clinical benefit over sequential treatment with single agents?
What new small molecules and antibodies are
effective against kidney cancer?
What treatments are best for nonclear cell
histologies?
697
698
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
57.
699
700
70.
71.
72.
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
701