06 - radiol clin n am 2007 - imaging of kidney cancer

29
Imaging of Kidney Cancer Jingbo Zhang, MD*, Robert A. Lefkowitz, MD, Ariadne Bach, MD Kidney cancers account for about 3% of all cancer cases as well as about 3% of all cancer deaths in the United States, with 38,890 new diagnoses and 12,840 deaths expected in 2006 [1]. According to the National Cancer Institute’s Surveillance, Epide- miology, and End Results (SEER) Program, between 1998 and 2002, the age-adjusted incidence and death rates for kidney cancers were 12.1 and 4.2 per 100,000, and between 1995 and 2001, the over- all 5-year survival rate of patients with kidney can- cer was 64.6% [2]. It is estimated that approximately $1.9 billion is spent in the United States each year on treatment of kidney cancer [3]. The incidence of kidney cancers has been increasing at a rate of about 2% per year for the past 30 years [2]. The overall mortality rate from kidney cancers has increased slightly over the past 2 decades, but not as rapidly as the incidence rate. This discrep- ancy is due to a significant improvement in 5-year survival [4]. These trends may be at least partially at- tributable to improved diagnostic capabilities. Newer radiographic techniques are detecting renal tumors more frequently and at a lower disease stage, when tumors can be resected for cure [5–11]. The kidney is a retroperitoneal structure sur- rounded by perirenal fat and the renal (Gerota’s) fascia. Most renal tumors arise from the renal pa- renchyma (referred to as renal cell tumors, renal cortical tumors, or renal parenchymal tumors), with a much smaller number arising from the uro- thelium of the renal collecting system (urothelial carcinoma or transitional cell carcinoma [TCC]) or the mesenchyma (eg, angiomyolipoma, leio- myoma, liposarcoma). Advances in molecular ge- netics in the last decade have expanded understanding of renal cell tumors significantly. Now it is understood that renal cortical tumors are a family of neoplasms with distinct cytogenetics and molecular defects, unique histopathologic fea- tures, and different malignant potentials [12–16]. In the conclusions of a workshop entitled ‘‘Impact of Molecular Genetics on the Classification of Renal Cell Tumours,’’ which was held in Heidelberg in Oc- tober 1996, a new classification system was pro- posed [14]. This classification system subdivides renal cell tumors into benign and malignant paren- chymal neoplasms and, where possible, limits each subcategory to the most commonly documented RADIOLOGIC CLINICS OF NORTH AMERICA Radiol Clin N Am 45 (2007) 119–147 Department of Radiology, Memorial Sloan-Kettering Cancer Center, Cornell University Weill Medical College, 1275 York Avenue, C278D, New York, NY 10021, USA * Corresponding author. E-mail address: [email protected] (J. Zhang). - Detection and diagnosis - CT scan Solid renal tumors Cystic renal tumors - MR imaging - Ultrasonography - Nuclear scintigraphy - Other renal tumors Urothelial carcinoma Metastases Lymphoma Primary renal mesenchymal tumors - Staging - Treatment planning - Imaging follow-up - Summary - References 119 0033-8389/07/$ – see front matter ª 2006 Elsevier Inc. All rights reserved. doi:10.1016/j.rcl.2006.10.011 radiologic.theclinics.com

Upload: others

Post on 11-Sep-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

R A D I O L O G I CC L I N I C S

O F N O R T H A M E R I C A

Radiol Clin N Am 45 (2007) 119–147

119

Imaging of Kidney CancerJingbo Zhang, MD*, Robert A. Lefkowitz, MD, Ariadne Bach, MD

- Detection and diagnosis- CT scan

Solid renal tumorsCystic renal tumors

- MR imaging- Ultrasonography- Nuclear scintigraphy- Other renal tumors

Urothelial carcinoma

MetastasesLymphomaPrimary renal mesenchymal tumors

- Staging- Treatment planning- Imaging follow-up- Summary- References

Kidney cancers account for about 3% of all cancercases as well as about 3% of all cancer deaths in theUnited States, with 38,890 new diagnoses and12,840 deaths expected in 2006 [1]. According tothe National Cancer Institute’s Surveillance, Epide-miology, and End Results (SEER) Program, between1998 and 2002, the age-adjusted incidence anddeath rates for kidney cancers were 12.1 and 4.2per 100,000, and between 1995 and 2001, the over-all 5-year survival rate of patients with kidney can-cer was 64.6% [2]. It is estimated thatapproximately $1.9 billion is spent in the UnitedStates each year on treatment of kidney cancer [3].The incidence of kidney cancers has been increasingat a rate of about 2% per year for the past 30 years[2]. The overall mortality rate from kidney cancershas increased slightly over the past 2 decades, butnot as rapidly as the incidence rate. This discrep-ancy is due to a significant improvement in 5-yearsurvival [4]. These trends may be at least partially at-tributable to improved diagnostic capabilities.Newer radiographic techniques are detecting renaltumors more frequently and at a lower diseasestage, when tumors can be resected for cure [5–11].

0033-8389/07/$ – see front matter ª 2006 Elsevier Inc. All righradiologic.theclinics.com

The kidney is a retroperitoneal structure sur-rounded by perirenal fat and the renal (Gerota’s)fascia. Most renal tumors arise from the renal pa-renchyma (referred to as renal cell tumors, renalcortical tumors, or renal parenchymal tumors),with a much smaller number arising from the uro-thelium of the renal collecting system (urothelialcarcinoma or transitional cell carcinoma [TCC])or the mesenchyma (eg, angiomyolipoma, leio-myoma, liposarcoma). Advances in molecular ge-netics in the last decade have expandedunderstanding of renal cell tumors significantly.Now it is understood that renal cortical tumorsare a family of neoplasms with distinct cytogeneticsand molecular defects, unique histopathologic fea-tures, and different malignant potentials [12–16].In the conclusions of a workshop entitled ‘‘Impactof Molecular Genetics on the Classification of RenalCell Tumours,’’ which was held in Heidelberg in Oc-tober 1996, a new classification system was pro-posed [14]. This classification system subdividesrenal cell tumors into benign and malignant paren-chymal neoplasms and, where possible, limits eachsubcategory to the most commonly documented

Department of Radiology, Memorial Sloan-Kettering Cancer Center, Cornell University Weill Medical College,1275 York Avenue, C278D, New York, NY 10021, USA* Corresponding author.E-mail address: [email protected] (J. Zhang).

ts reserved. doi:10.1016/j.rcl.2006.10.011

Page 2: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al120

genetic abnormalities. Historically, malignant renalcell tumors have been described as a single entity,such as hypernephroma, renal cancer, renal adeno-carcinoma, and renal cell carcinoma (RCC). By theHeidelberg classification, malignant renal paren-chymal tumors include common or conventionalRCC (also known as the clear cell type, accountingfor about 75% of renal neoplasms in surgical se-ries), papillary RCC (accounting for 10%), chromo-phobe RCC (accounting for 5%), collecting duct(Bellini duct) carcinoma (accounting for 1%), andrarely unclassified tumors [14]. Medullary carci-noma of the kidney is a variant of collecting ductcarcinoma and was initially described in patientswho were sickle cell trait–positive [14]. Benign re-nal parenchymal tumors include renal oncocytoma(5%) and the rarer metanephric adenoma, meta-nephric adenofibroma, and papillary renal celladenoma.

Malignant renal cell tumors occur nearly twice asoften in men as in women [9]. The age at diagnosisis generally older than 40 years; the median age is inthe mid-60s [9]. Past international, multicenter,population-based, case-control studies have pro-vided insight into the environmental risk factorsfor development of malignant renal cell tumor[17–23]. Diet [23]; cigarette smoking [20]; unop-posed estrogen exposure [17]; body mass index inwomen and, to a lesser extent, in men [21]; occupa-tional exposure to petroleum products, heavymetals, or asbestos [18]; and hypertension, treat-ment for hypertension, or both [19] have been re-ported associated with malignant renal celltumors. The risk of RCC also has been reported tobe increased in patients with acquired cystic kidneydisease associated with long-term hemodialysis[24]. Heredity plays a role in some cases, with therisk of the disease increasing fourfold when a first-degree relative carries the diagnosis [22], but onlya small fraction of patients have an affected familymember. Certain familial syndromes, such as vonHippel–Lindau disease [26], also are associatedwith malignant renal cell tumors. In addition,a few kindreds with familial clear cell carcinomahave been reported and were found to have chro-mosomal abnormalities [14,27,28]. Cytogeneticand molecular genetic analyses of tumors carriedby these families have contributed substantially tounderstanding of renal tumor pathogenesis ona molecular level [29].

The different subtypes of renal cell tumors areassociated with distinctively different disease prog-ression and metastatic potential [30–33]. Theconventional clear cell carcinomas, accounting forapproximately 90% of the metastases, have thegreatest metastatic potential, whereas papillaryand chromophobe carcinomas (accounting for

approximately 10% of metastases) are associatedwith less metastatic potential [33]. The overall5-year survival rates for papillary and chromophobesubtypes (80–90%) are much higher than that forconventional RCC (50–60%) [34–36]. Amongthese three most common types of malignant renalcortical tumors, the chromophobe type is associ-ated with the least metastatic potential and bestprognosis [12,15,30,31,33,36,37].

In addition to histologic subtypes, other inde-pendent predictors of patient prognosis include pa-tient age, functional status, symptomatic tumorpresentation, stage (discussed in detail later in thisarticle), and the Fuhrman nuclear grade [38–47].Symptomatic tumor presentation, higher stage,and higher nuclear grades correlate with greater bi-ologic aggressiveness of the tumor and increasedmetastatic potential.

Bilateral multifocal renal tumors are present inapproximately 5% of patients with sporadic renaltumors [48,49]. As in solitary disease, conventionalclear cell histology is the most common histologicsubtype; concordance between histologic subtypesamong tumors was found to be 76% in one study[48]. Bilateral multifocal tumors and unilateral tu-mors have comparable prognoses when treatedwith surgery [48,49].

Detection and diagnosis

Common symptoms that lead to the detection ofa renal mass are hematuria, flank mass, and flankpain. The combination of these symptoms is pres-ent, however, in only 10% of cases. Less frequently,patients present with signs or symptoms resultingfrom metastatic disease, such as bone pain, adenop-athy, and pulmonary symptoms. Other presenta-tions include signs or symptoms such as fever,weight loss, anemia, or a varicocele.

Diagnostic imaging of renal masses has evolveddramatically over the past 2 decades. CT, MR imag-ing, and ultrasonography are being increasinglyperformed in place of traditional diagnostic imag-ing tests, such as intravenous urography and angi-ography. Among these diagnostic tools, CT isconsidered the modality of choice for detectionand diagnosis of renal cortical tumors, with MRimaging and ultrasound frequently applied as prob-lem-solving tools or in patients with contraindica-tions to contrast-enhanced CT. The advent ofmultislice CT has led to faster acquisition times,higher spatial resolution, and the greatest numberof CT examinations ever performed; this has ledto a great increase in the detection and earlier diag-nosis of renal cortical tumor [5–11]. Currently, upto 70% of tumors are discovered incidentally, witha median tumor size of less than 5 cm

Page 3: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 121

[10,11,16,50]. Among all renal cortical tumors de-tected, 20% may be benign, and 25% may be rela-tively indolent papillary or chromophobecarcinomas [30,31,34–36].

Although it is important to make a preoperativediagnosis of the tumor type for treatment planningand patient counseling, there are no well-estab-lished imaging criteria for diagnosing the histologicsubtypes without operative resection, and the roleof biopsy is controversial [51,52]. It has beensuggested that certain imaging features may beassociated with different renal cortical tumorsubtypes [53–58]. The diagnosis of renal masses,especially the incidentally detected ones, remainsproblematic, however [5,59,60]. The techniquesand established and potential roles of imaging inthe diagnosis of renal cortical tumors with solidsoft tissue components are presented subsequently.A discussion of imaging of cystic renal massesfollows.

CT scan

A dedicated imaging protocol is needed for opti-mized evaluation of the renal mass by CT. At the au-thors’ institution, a CT scan dedicated forevaluation of a renal mass typically consists of threeimaging series performed during breath hold: pre-contrast, corticomedullary phase, and late nephro-graphic/early excretory phase [61–63]. Precontrastimages are essential for evaluation of the presenceof calcifications and provide a baseline densitymeasurement for evaluating the degree and patternof enhancement in cystic or solid renal masses [63].Corticomedullary images (typical scan delay 70–85seconds after injection) are superior in the assess-ment of lesion vascularity, renal vascular anatomy,and tumor involvement of venous structures [61].In addition, they are probably the most informativeimages for lesion characterization. A relatively thinslice thickness of 2.5 mm may be obtained throughthe kidneys. To minimize radiation exposure, a stan-dard slice thickness of 5 mm may be obtainedthrough the rest of the body if such imaging is tobe performed in the same setting. Not all renal tu-mors are well delineated during the corticomedul-lary phase, however, and images obtained duringa later phase of enhancement (ie, the nephro-graphic or excretory phase) must be included to fa-cilitate the detection of renal masses, especiallythose of smaller size [63–70]. In addition, excretoryphase images (these typically can be achieved witha scan delay of 3 minutes) are helpful for delinea-tion of anatomic abnormalities or tumor involve-ment of the renal collecting system [61]. Highaccuracy (sensitivity 100%, specificity 95%) has

been reported in the detection of renal masseswhen proper technique is applied [70].

Solid renal tumors

In terms of differentiation of solid renal tumors,limited investigations have been performed in thepast regarding whether certain imaging featuresmay be associated with specific subtypes of renalcortical tumor [53–58]. The most consistent andvaluable parameter probably is the degree of en-hancement because clear cell carcinomas enhanceto a greater degree than other subtypes of malignantlesions, especially papillary carcinomas. This wasshown in studies from the groups of Herts, Jinzaki,Kim, and Ruppert-Kohlmayr [53–55,57,58]. In ad-dition, Herts and colleagues [53] showed that pap-illary RCCs are typically homogeneous, and Sheirand associates [58] showed that cystic degenerationwas more evident in the clear cell subtype than inthe other subtypes. Sheir and associates [58] re-ported, however, that a hypervascular pattern wasmore prevalent in papillary than in chromophobesubtypes, a finding inconsistent with the findingsof the other studies cited. Most of these studies in-cluded only malignant lesions or in some cases sub-groups of malignant lesions in their analyses.

In a series of 198 solid renal tumors that were re-sected at the authors’ institution, 55% were of theclear cell type, 15% were papillary, 12% were chro-mophobe, 7% were oncocytomas, and 3% werelipid-poor angiomyolipomas [71]. Almost half ofall renal tumors are either benign or relatively indo-lent malignant tumors. Conventional clear cell renalcarcinoma is the most vascular type among all ma-lignant renal cortical tumors, as shown by its greaterdegree of enhancement after administration of in-travenous contrast material [54,55,58,71]. A mixedenhancement pattern containing enhancing solidsoft tissue and low-attenuation areas that may rep-resent cystic or necrotic changes was most predic-tive of the clear cell type (Fig. 1) [71].Oncocytomas may overlap, however, with clearcell RCC in terms of imaging features and degreeof enhancement (Fig. 2) [54,71]. Classic angio-graphic findings for oncocytoma have been reportedin the past, including a spoke-wheel pattern, a ho-mogeneous tumor blush, and a sharp, smoothrim [72]. None of these findings is specific, how-ever, and a RCC may have any or all of the classicfindings [72]. On CT scans, the diagnosis of oncocy-toma may be suggested if a central stellate scar isidentified within an otherwise homogeneous tu-mor (Fig. 3) [73]. Oncocytoma may often manifestas a complex hypervascular mass, however, some-times associated with adjacent neovascularity andperinephric stranding, and cannot be reliably differ-entiated from clear cell carcinoma (Fig. 4) [71].

Page 4: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al122

In the authors’ study, papillary RCCs were typi-cally less vascular compared with most other typesof renal tumors and most commonly manifestedas homogeneous (Fig. 5) or peripheral enhance-ment (Fig. 6) [71]. A low tumor-to-aorta enhance-ment ratio or tumor-to-normal renal parenchymaenhancement ratio was highly indicative of papil-lary RCC [71]. This is consistent with the findingof Herts and colleagues [53] that a high tumor-to-parenchyma enhancement ratio R25%) essentiallyexcludes the possibility of a tumor being papillaryRCC.

Chromophobe RCCs are more variable in their de-grees and patterns of enhancement (Fig. 7) [71].

Fig. 1. (A) A 50-year-old man with clear cell carcinomain right kidney. Contrast-enhanced CT image duringrenal parenchymal phase shows a right renal masswith a mixed enhancement pattern containing en-hancing solid soft tissue and low-attenuation areasthat may represent cystic or necrotic changes. (B) A63-year-old man with clear cell carcinoma in left kid-ney. Contrast-enhanced CT image during renal paren-chymal phase shows a left renal mass with a mixedenhancement pattern with a greater amount of solidcomponents than (A).

Homogeneously high-attenuation on unenhancedCT images and homogeneous enhancement oncontrast-enhanced CT images have been reportedto suggest angiomyolipoma containing abundantmuscle and minimal fat [74]. Based on the authors’experience, however, these features may be sharedby fat-poor angiomyolipomas and the more indo-lent type of RCCs (papillary and chromophobe)(Fig. 8) [71].

Generally, the presence of calcifications in a solidrenal mass suggests malignancy [71,75]. Rarely,

Fig. 2. A 56-year-old woman with right renal oncocy-toma. Contrast-enhanced CT image during renal pa-renchymal phase shows a right renal mass witha mixed enhancement pattern that mimics clear cellRCC.

Fig. 3. A 58-year-old man with left renal oncocytoma.Contrast-enhanced CT image during renal parenchy-mal phase shows a left renal mass with nearly homo-geneous enhancement and a small low-attenuationcentral scar.

Page 5: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 123

a malignant renal tumor may be diffusely calcified(Fig. 9).

Cystic renal tumors

Numerous articles have been published on the sub-ject of cystic renal masses. The widely quoted Bos-niak classification system grades the cystic renalmasses for the likelihood of malignancy based onthe complexity of these lesions [76]. When anysolid enhancing component is present, the cystic re-nal mass is graded as a Bosniak type 4 lesion, highlysuspicious for malignancy. As discussed earlier, fur-ther differentiation of this group of lesions may bepossible based on the characteristics of their solidsoft tissue components. Cystic tumors with thinwalls and septations without solid components inan adult may represent benign cystic nephroma,multilocular cystic RCC, or, rarely, cystic hamarto-ma of the renal pelvis [31]. These tumors are similarin their gross appearances and cannot be differenti-ated by preoperative imaging studies (Fig. 10) [31].When such a cystic mass does represent multilocu-lar cystic RCC, however, it may be of no, or at mostlittle, malignant potential [31] and carries a dis-tinctly better prognosis than other forms of RCC[77]. This is probably because although thesemasses may contain clear cells, they generally are as-sociated with a lower Fuhrman grade (1 or 2) onhistopathology.

One common pitfall in characterizing renal le-sions by CT is the presence of pseudoenhancementin renal cysts on contrast-enhanced CT images. Thispseudoenhancement is thought to be due to vol-ume averaging and beam hardening effects, and

Fig. 4. A 94-year-old woman with left renal oncocyto-ma. Contrast-enhanced CT image during renal paren-chymal phase shows a 7-cm left renal mass. The massshows a degree of enhancement similar to that of re-nal parenchyma with heterogeneity and peritumoralvascularity mimicking clear cell RCC.

the degree of pseudoenhancement is greater insmaller renal cysts [78–84]. In certain cases, thedegree of pseudoenhancement may well surpass the10 HU threshold commonly used to differentiatenonenhancing simple cysts from enhancing solidlesions (Fig. 11) [78–84]. In these cases, ultrasoundor MR imaging may prove helpful.

MR imaging

MR imaging has many advantages over other mo-dalities in the detection and staging of renal neo-plasms, owing to its intrinsic high soft tissuecontrast, direct multiplanar imaging capabilities,and availability of a non-nephrotoxic, renallyexcreted contrast agent [85]. State-of-the-art MR im-aging of renal masses includes the following breath-hold sequences: (1) a T1-weighted in and out ofphase gradient echo sequence, which is helpful in

Fig. 5. A 63-year-old man with papillary carcinoma ofthe right kidney. (A) Precontrast CT image showsa mass of smooth counter and homogeneous attenu-ation the same as or slightly higher than that of theadjacent renal parenchyma. (B) Postcontrast CT imagein the renal parenchymal phase shows uniform, mildenhancement of 35 HU in the renal mass.

Page 6: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al124

identification of macroscopic and microscopic fatin a renal tumor (Fig. 12) [86]; (2) a T2-weightedhalf-Fourier single-shot fast spin-echo sequence inaxial or coronal planes, which is useful for evaluat-ing the overall anatomy, renal collecting system,and complexity of a cystic renal lesion (Fig. 10);and (3) a dynamic contrast-enhanced T1-weightedfat-suppressed sequence [87]. For dynamic con-trast-enhanced images, three-dimensional fastspoiled gradient echo sequences are typically per-formed [87–89] before and after contrast adminis-tration during the arterial, corticomedullary, andnephrographic phases for evaluation of the pres-ence and pattern of enhancement in a renal mass[87,88]. Multiplanar reconstruction may be per-formed if necessary to delineate better the spatial re-lationship of the renal mass to adjacent anatomicstructures. If necessary, a dedicated MR angiographysequence during the arterial phase may be per-formed for better visualization of accessory renalvessels and facilitation of surgical planning. Coro-nal T1-weighted images also may be obtained dur-ing the excretory phase with administration ofdiuretics, from which maximum intensity projec-tion images can be obtained to produce intrave-nous pyelography–like images.

MR imaging is useful in the detection and differ-entiation of cystic and solid renal lesions [90], withaccuracy comparable or superior to that of CT [91].In addition, MR imaging can be performed safely inpatients with renal failure and used for evaluationof renal tumors in these patients [92,93], an obvi-ous advantage because many patients with renal

Fig. 6. A 58-year-old man with a 15-cm papillary RCCarising from the right kidney. Contrast-enhanced CTimage during renal parenchymal phase shows a rightrenal mass with mild enhancement of 35 HU in pe-ripherally distributed soft tissue. No enhancement isidentified in the central low-attenuation region.

masses are at risk of renal insufficiency at presenta-tion or after surgical treatment. For these reasons,MR imaging may function as an excellent tool forinitial diagnosis and post-treatment follow-up inthese patients. It is reliable for evaluation of smallrenal masses [92,94] owing to its superior soft tis-sue contrast, whereas CT can be problematic be-cause of pseudoenhancement (Fig. 11). Inaddition, because of its multiplanar capability, MRimaging may be superior to CT for determiningthe origin of a renal mass [71].

Solid renal tumors are typically isointense orslightly hypointense on T1-weighted images[90,95], although some renal tumors may containhemorrhage or a lipid component and show T1

Fig. 7. (A) A 60-year-old woman with left chromo-phobe RCC. Contrast-enhanced CT image during re-nal parenchymal phase shows a left renal mass withhomogeneous enhancement. (B) A 48-year-oldwoman with right chromophobe RCC. Contrast-enhanced CT image during renal parenchymal phaseshows a right renal mass with heterogeneousenhancement.

Page 7: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 125

Fig. 8. A 58-year-old woman with lipid-poor right re-nal angiomyolipoma. (A) Precontrast CT image showsa homogeneous soft tissue mass that has slightlyhigher attenuation than the renal parenchyma. (B)Postcontrast parenchymal phase CT image shows ho-mogeneous enhancement of approximately 120 HUin this mass. Note peripheral location of the mass.

hyperintensity (Fig. 12) [93]. Clear cell carcinomasmay contain intracellular lipids and show focal ordiffuse signal loss on opposed-phase images, whichdoes not always indicate angiomyolipoma (seeFig. 12) [87,96,97]. Renal cortical tumors tend tobe mildly hyperintense [95] on T2-weighted imagesand show variable enhancement on dynamic con-trast-enhanced images [90]. Simple cysts are hypo-intense on T1-weighted images and hyperintenseon T2-weighted images. Although some complexcysts may show a higher T1 signal and lower T2 sig-nal owing to hemorrhage, debris, or proteinaceousmaterial, there should be no enhancement in cystsafter administration of contrast. Identification ofthe presence of contrast enhancement is essentialin diagnosing a solid renal neoplasm. It has beenreported that the optimal percentage of enhance-ment threshold for distinguishing cysts from solidtumors on MR imaging is 15% when measurementis performed 2 to 4 minutes after administration ofcontrast material [98]. This threshold may beachieved with quantitative analysis of enhancementwith signal intensity measurements [98]. Qualita-tive analysis of enhancement with image subtrac-tion is equally accurate, particularly in the settingof masses that are hyperintense on unenhancedMR images [99]. Similar to those seen on CT, threepatterns of enhancement have been observed inRCCs on MR imaging: predominantly peripheral,heterogeneous, and homogeneous [90].

CT and MR imaging perform similarly in classify-ing most cystic renal masses [100]. In some cases,however, MR images may depict additional septa,thickening of the wall or septa, or enhancement,which may lead to an upgraded Bosniak cyst classi-fication and affect case management [100].

Fig. 9. A 59-year-old man with papillary renal carcinoma. (A) Ultrasound of the right kidney shows a 5-cm cal-cified mass in the upper pole. (B) Unenhanced CT image shows heavy calcification in the periphery of this mass.

Page 8: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al126

Fig. 10. (A) A 68-year-old woman with breast cancer.T2-weighted single-shot fast spin-echo MR imageshows a complex cystic mass in the left kidney, whichwas proved to be a multilocular cystic nephroma onpathology. (B) A 52-year-old woman with multilocu-lar cystic RCC in the left kidney. Contrast-enhancedCT image at renal parenchymal phase shows a cysticmass containing several thickened enhancing septa-tions in the left kidney. Pathology revealed a Furhmannuclear grade of I/IV.

Ultrasonography

Ultrasound is an important problem-solving toolfor evaluation of renal masses. One of the most im-portant roles of ultrasound is in the characteriza-tion of renal lesions as cystic or solid. Because thediagnosis of a simple renal cyst denotes benignity,strict criteria need to be adhered to. The lesionmust be completely anechoic, have a thin imper-ceptible wall, have posterior enhancement, havea round or oval shape, and be avascular. Rarely, a be-nign cyst can become complex in the setting ofhemorrhage or infection and could mimic a solidlesion (Fig. 13). Cysts that do not fulfill all of thecriteria of a simple cyst are complex, and the possi-bility of a cystic renal carcinoma may need to beconsidered depending on a cyst’s radiographic fea-tures (Fig. 14). Correlation with other imagingmodalities, such as CT and MR imaging, is recom-mended. Features on ultrasound suggesting a malig-nant cystic lesion include a thickened cystic wall,numerous septations, thickened or nodular septa-tions, irregular or central calcifications, and thepresence of flow in the septations or cystic wall onDoppler imaging [101].

Most RCCs on ultrasound are solid. Cystic areasand calcifications may be present (Fig. 15). Small,3 cm or less, renal masses are more likely to be hy-perechoic than larger tumors [102]. Ultrasound andcross-sectional imaging modalities (CT and MR im-aging) complement each other in the characteriza-tion of renal lesions. In the subgroup of patientswhose renal lesions are ‘‘indeterminate’’ on ultra-sound, a dedicated renal protocol CT or MR imag-ing may help characterize the lesion further [103].Conversely, ultrasound may prove useful for renallesions that are considered indeterminate on CT[95].

Solid renal masses on ultrasound usually need tobe evaluated with a renal protocol CT for the

Fig. 11. A 58-year-old man with benign high-density cyst in left kidney. (A) Precontrast CT image shows a smallleft renal mass of 71 HU in attenuation. (B) Contrast-enhanced CT image during renal parenchymal phase showsincreased attenuation to 94 HU (a pseudoenhancement of 23 HU) in this small mass.

Page 9: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 127

Fig. 12. A 67-year-old woman with right renal clear cell carcinoma. (A and B) T1-weighted dual-echo gradientecho MR images of the right kidney show loss of signal in right renal mass on opposed phase relative to in-phaseimage. (A) In-phase. (B) Opposed phase.

presence of fat. If fat is present in the lesion on CT,in most cases an angiomyolipoma can be diag-nosed. Rarely, however, RCC may engulf the perire-nal or sinus fat, and a liposarcoma may containfatty components. Malignancy should be suspectedon the basis of the following criteria: presence of in-tratumoral calcifications; large, irregular tumor in-vading the perirenal or sinus fat; large necrotictumor with small foci of fat; and association withnonfatty lymph nodes or venous invasion [104–106].

Limited attempts have been made to differentiaterenal cancer into the different histologic subtypesbased on ultrasound characteristics. PapillaryRCCs tend to be hypoechoic or isoechoic, butsome also may be hyperechoic (Fig. 16) [56].Work has been done in ultrasound using Dopplerimaging, which allows for assessment of vascularflow [107]. Contrast-enhanced ultrasound has

Fig. 13. A 67-year-old woman with benign epithelialcyst in left kidney. Ultrasound of the left kidneyshows a small lesion (arrow) with internal echoesmimicking a solid lesion.

been found to be useful in the diagnosis of renalcortical tumors and in the detection of tumor bloodflow in hypovascular renal masses [108,109]. Astudy from the authors’ institution indicates thatvascular flow within a renal mass, identified bycolor and power Doppler, is strongly associatedwith conventional clear cell carcinoma [110]. Thevascular distribution at power Doppler potentiallycould add information in differentiation of smallsolid renal masses. It has been shown that periph-eral or mixed penetrating and peripheral patternsare seen in all RCCs and some benign angiomyoli-pomas and oncocytomas, whereas intratumoral fo-cal or penetrating patterns are characteristic ofangiomyolipoma [111].

Renal ultrasound is not considered a usefulscreening modality because small lesions can beeasily missed [112]. CT detects more and smaller re-nal masses than does ultrasound, but the two

Fig. 14. A 40-year-old man with clear cell renal carci-noma of the right kidney. Ultrasound revealed a 5-cmcomplex cystic mass in the lower pole of the right kid-ney. Color Doppler documented the presence of vas-cularity in the septations (not shown).

Page 10: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al128

modalities are comparable in characterizing 1- to 3-cm lesions [113]. Although renal sonography maynot be the best method for generalized primaryscreening, it still may be beneficial in ‘‘secondary’’screening in a more selected patient population,such as in the elderly asymptomatic population[114].

Fig. 15. (A) A 74-year-old man with clear cell renal car-cinoma in right kidney. Ultrasound showed a 4 cm � 5cm heterogeneous mass in the posterior medial rightkidney containing solid and cystic components. (B) A37-year-old man with clear cell renal carcinoma of theleft kidney. Ultrasound showed a 6 cm � 7 cm mass inthe interpolar region of left kidney. The mass waspredominately solid with scattered cystic areas.

Fig. 16. A 46-year-old man with papillary renal carci-noma in right kidney. Ultrasound showed a solid5-cm hyperechoic mass in the lower pole of the rightkidney.

Nuclear scintigraphy

Although fluorodeoxyglucose (FDG) positronemission tomography (PET) has been establishedas an efficient imaging modality for the manage-ment of certain cancers, its role in imaging of renalcortical tumors has not yet been clearly defined.Varying sensitivity of FDG PET for the detectionof renal malignancy has been reported, rangingfrom 40% to 94% [115–118]. Several factors mayexplain the false-negative results of FDG PET fordetection of renal malignancy. First, normal renalexcretion of FDG may have decreased the contrastbetween tumor and normal surrounding tissues,reducing the efficacy of PET in detecting primaryrenal malignancy [115]. Second, other factors,such as histologic subtypes, Fuhrman grades, ortumor vascularity, may have played a role. False-positive results by FDG PET have been reported inpatients with benign inflammatory processes ofthe kidney or benign tumors such as oncocytomas[115,119]. A discrepancy has been observedbetween poor visualization of the primary renalmalignancy and high uptake of FDG by metastasesin the same patient [115]. FDG PET may have a po-tential role in evaluation of distant metastases, es-pecially when equivocal findings are present onconventional studies, and in the differentiation be-tween recurrence and post-treatment changes[115,117,118,120]. PET with new radiopharmaceu-tical agents, such as radioisotope-labeled monoclo-nal antibodies with specificity for RCC, may be ableto provide in vivo diagnosis and determination ofphenotype in the future.

Other renal tumors

Urothelial carcinoma

Urothelial tumors of the renal pelvis representapproximately 7% of primary renal neoplasms[121] and approximately 5% of urinary tract tumors[122]. Urothelial tumors of the renal pelvis are sim-ilar to those in the urinary bladder in pathologicfeatures, epidemiologic distributions, and risk fac-tors (which are discussed in detail in the article byZhang and colleagues elsewhere in this issue). Uro-thelial carcinoma (or TCC) accounts for 90% of pel-vicalyceal malignant tumors, whereas squamouscell carcinomas, which are radiologically indistin-guishable from TCC, account for the other 10%[122].

TCC is often multifocal, presenting as synchro-nous or metachronous lesions. This known multi-focality requires thorough examination of theentire urinary tract in high-risk patients, such as pa-tients with bladder cancer or known chemical

Page 11: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 129

exposure [121]. Traditionally, excretory intravenousurography (IVU) has been the screening examina-tion of choice for the detection of upper urinarytract neoplasms for patients with hematuria or forhigh-risk patients. IVU has been shown to miss40% of upper tract neoplasms, however [121].With the advent of multidetector CT enabling therapid acquisition of thin-section images throughthe entire urinary tract, CT urography (CTU) has be-gun to supersede IVU as the imaging examinationof choice to detect upper urinary tract tumors. Thetechniques and applications of CTU are discussedin detail in the article by Zhang and colleagues else-where in this issue.

On imaging, TCC of the renal pelvis manifests asa focal soft tissue mass or focal wall thickening. Onnoncontrast images, these tumors measure approx-imately 30 HU and most of the time can be differ-entiated from water (approximately 0 HU), bloodclot (50–75 HU), and calculi (>100 HU) [123].The enhancement pattern of urothelial neoplasms

(typically mild-to-moderate early enhancementwith washout on delayed images) can be similarto that of renal cortical neoplasms, although the lat-ter are typically more vascular [123–125]. Of TCCs,85% are early stage, superficial papillary neoplasms.In the excretory phase of CTU, early stage TCCappears as a sessile filling defect within the high-density contrast excreted into the renal collectingsystem; the tumor expands the collecting systemwith compression of the renal sinus fat (Fig. 17).Alternatively, early stage tumors can appear as focalor diffuse mural thickening, pelvicalyceal irregular-ity, or focally obstructed calyces (Fig. 18)[123,124]. Fifteen percent of TCCs are more aggres-sive, infiltrating lesions that show mural thickeningand present at a more advanced stage, often with in-vasion into the renal parenchyma (Fig. 19) [124].

CT plays a role in staging urothelial tumors. Al-though CT cannot accurately distinguish T1 tumor(limited to uroepithelium and lamina propria)from T2 tumor (extending into muscularis propria,

Fig. 17. A 76-year-old woman with noninvasive papillary TCC of left renal pelvis. (A) Noncontrast CT image showssoft tissue in left renal pelvis (arrow) slightly higher in attenuation than renal parenchyma. (B) Contrast-enhanced image during renal parenchymal phase shows uniform enhancement of approximately 80 HU inthe mass. A clear fat plane is identified between the tumor and the kidney, indicating absence of renal paren-chymal invasion. (C and D) Delayed, excretory phase CT images show a corresponding filling defect in thecollecting system on soft tissue (C) and bone windows (D).

Page 12: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al130

but not beyond), this modality is accurate in distin-guishing early stage, T1 and T2 tumors from ad-vanced-stage, T3 (invading peripelvic fat or renalparenchyma) and T4 (invading adjacent organs orabdominal wall or extending through renal paren-chyma into perinephric fat) tumors; this distinctionis important because studies have shown a signifi-cant difference in survival between stage II and IIIlesions [125]. Advanced stage tumor is suspectedwhen the normally low-density renal sinus fatshows an increase in attenuation owing to tumorinfiltration, or when the fat plane between the tu-mor and the renal parenchyma is lost; in somecases, CT may show direct invasion of tumor into

Fig. 18. A 54-year-old man with high-grade papillaryurothelial carcinoma of left renal pelvis confined tomucosa of renal pelvis (stage T1). Contrast-enhancedCT shows a uniform soft tissue mass in left renal pelvisencasing a renal stent (arrow). A thin fat plane ismaintained around the mass.

the renal parenchyma, which can appear as an infil-trating renal mass (Fig. 19) [123–125]. Occasion-ally, uroepithelial neoplasms invading the renalparenchyma can appear similar radiologically tocentrally located renal cortical neoplasms invadingthe renal sinus, although in general TCC tends to bemore centrally located and is more likely to expandthe kidney centrifugally, while preserving the renalcontour (Fig. 20) [122,125]. Although advancedTCC can invade the renal vein and inferior venacava on rare occasions, RCC has a much greater pre-dilection for spreading in this fashion [125]. Forstaging of advanced tumors, CT, superior to IVU,also allows high-quality imaging of the lymph no-des, liver, bones, and lungs, which are the mostcommon sites of metastatic disease from urothelialneoplasms [124].

In the past, radical nephroureterectomy has beenthe standard treatment for renal and ureteral TCC.Newer renal-preserving techniques, such as laser ab-lation, endoscopic fulguration, or segmental exci-sion, are now the preferred treatments, however,for patients who have early stage, low-grade tumors,a solitary kidney, bilateral TCC, or suboptimal renalfunction and for patients who are poor surgical can-didates [123,124]. The clinical outcomes for thesenewer procedures are similar to outcomes formore radical surgery when the tumors are detectedat an early stage [124]. Early detection of upper tractTCC with urine cytology and cross-sectional imag-ing is essential.

Metastases

Rarely, solid tumors—most commonly primary tu-mors of the lung, breast, stomach, or contralateralkidney—do metastasize to the kidney [87,126]. Re-nal metastases are often multiple and bilateral andusually are seen with metastases elsewhere in the

Fig. 19. A 71-year-old woman with invasive TCC of the left renal pelvis. (A and B) CT urogram shows a mass in therenal pelvis on the renal parenchymal (A) and excretory phase (B) images. The fat plane between the mass andthe renal parenchyma is obliterated (arrows), indicating tumor invasion into renal parenchyma as confirmed bypathology.

Page 13: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 131

Fig. 20. A centrally located renal cortical tumor may mimic a uroepithelial tumor with renal parenchymal inva-sion. (A) A 72-year-old woman with conventional clear cell carcinoma of the right kidney. Contrast-enhanced CTshows a right renal mass invading the renal sinus, mimicking advanced stage TCC of the renal pelvis. (B) A 69-year-old woman with squamous cell carcinoma of renal pelvis. Contrast-enhanced CT shows a heterogeneousmass in the renal sinus with renal parenchymal invasion.

body. In patients with known primary malignan-cies, renal metastases are four times more commonthan primary renal neoplasms [127]. In the absenceof metastases to other organs, however, a solitary re-nal mass in a patient with a known primary malig-nancy is more likely to represent primary RCC thanmetastatic disease [128]. The imaging features of re-nal metastases are otherwise nonspecific: Renal me-tastases are usually more infiltrative than RCC andless vascular, in particular, than conventional clearcell carcinoma; however, the imaging features ofRCC and renal metastases often overlap, and a bi-opsy is frequently necessary to distinguish these en-tities (Fig. 21) [87]. Because clinical management

Fig. 21. A 46-year-old woman with metastatic coloncancer. Contrast-enhanced CT image shows a solidmass in the right kidney proven to represent metasta-sis of colorectal origin at pathology.

for a primary renal tumor and a metastatic renallesion is completely different, it is important fora radiologist to consider this differential diagnosis.

Lymphoma

The kidney is one of the most common sites ofextranodal lymphoma. Most extranodal lympho-mas are non-Hodgkin lymphomas, and lymphomais the third most common tumor, after lung andbreast cancer, to involve the kidney secondarily[101,129]. Although 33% of patients with lym-phoma have renal involvement in autopsy studies,the ante mortem incidence ranges from 0.5% to8.3% [129].

Because lymphoid tissue is normally absent inthe kidney, it is believed that primary renal lym-phoma is extremely rare, with most cases secondar-ily involving the kidney by direct extension orhematogenous spread [101,129]. Direct extensionof lymphoma can occur when lymphoma of theretroperitoneum invades the renal capsule (trans-capsular spread) or infiltrates the renal sinus, encas-ing the ureter, renal pelvis, and renal vascularstructures [101]. Because of the lack of associated fi-brosis, lymphoma typically encases the renal hilarstructures without obstruction (Fig. 22) [101,129].From there, lymphoma can extend into the renalparenchyma via the medullary pyramids. Whenlymphoma gains access to the renal parenchyma,by direct extension or hematogenous spread, it ini-tially involves the renal interstitium, using vessels,collecting ducts, and nephrons as scaffolding togrow.

Because the cells infiltrate between and separate,rather than destroy, these structures, lymphoma canappear subtle on imaging studies in its early stages

Page 14: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al132

[101]. Lymphomatous lesions often cause relativelylittle mass effect and generally do not deform the re-nal collecting system or renal contour. As the le-sions enlarge, they may compress the calyces, butthe reniform shape of the kidney usually is pre-served even if the entire organ is infiltrated by tu-mor. Less often, these tumors may grow rapidly ina nonuniform fashion, creating a focal mass thatprojects beyond the contour of the kidney, mimick-ing a primary renal cortical neoplasm but remain-ing unencapsulated [101]. Renal lymphoma istypically homogeneous and shows a lower degreeof enhancement than adjacent renal parenchymaon CT and MR imaging. Renal lymphoma canhave several patterns on cross-sectional imaging[130]. Most cases manifest with multiple soft tissuemasses in one or both kidneys. Less common imag-ing patterns include a solitary renal mass, direct in-vasion of the renal hilum or renal sinus by bulkyretroperitoneal masses, and infiltration of the

Fig. 22. A 74-year-old man with small lymphocyticlymphoma. Contrast-enhanced CT image shows ho-mogeneous soft tissue infiltrating the renal sinusesbilaterally, encasing the renal hilar structures withouturinary obstruction or vascular occlusion (longarrows). Note associated retroperitoneal adenopathy(short arrow).

perirenal space without significant renal parenchy-mal involvement or diffuse infiltration of the entirekidney (Fig. 23) [129,131]. Most patients also pres-ent with adjacent retroperitoneal adenopathy (seeFigs. 22 and 23) [129].

Primary renal mesenchymal tumors

Primary renal mesenchymal tumors include malig-nant and benign lesions. Malignant renal mesen-chymal sarcomas are rare, comprising less than1% of all adult renal tumors and mostly involvingpatients between 40 and 70 years old [132,133].The most common primary renal sarcoma is leio-myosarcoma followed by liposarcoma and fibrosar-coma. Less common renal sarcomas includemalignant fibrous histiocytoma, rhabdomyosar-coma, osteosarcoma, chondrosarcoma, malignantperipheral nerve sheath tumor, clear cell sarcoma,and angiosarcoma [122,132,133].

Sarcomas of the kidney often arise from the renalcapsule or sinus and usually can be distinguishedfrom RCC because they lie outside the renal paren-chyma [122,133]. In contrast to RCC, renal sarco-mas are often extremely exophytic, displacing,distorting, or compressing the renal parenchymawithout parenchymal invasion; as a result, there isoften a smooth interface between the tumor andthe renal parenchyma [133,134]. Renal sarcomasthat do arise within the renal parenchyma are diffi-cult to distinguish from the more common renalcortical tumors, however. On CT, renal sarcomasare typically large heterogeneous soft tissue masseswith areas of necrosis (Fig. 24). A specific histologicdiagnosis usually cannot be made except in mostcases of liposarcoma, in which part or all of the tu-mor is composed of macroscopic fat. In contrast toRCC, renal sarcomas do not tend to invade therenal vein or inferior vena cava [133].

Fig. 23. A 51-year-old manwith mantle cell lymphomainvolving right kidney. (A)Contrast-enhanced CT showshomogeneous soft tissuemass infiltrating the perire-nal space without grossrenal parenchymal involve-ment or mass effect on the re-nal parenchyma. Noteretroperitoneal adenopathyin the inter-aortocaval region(arrow). (B) FDG PET scanshows intense uptake in theregion of the right kidney (ar-row) corresponding to thelymphomatous mass on CT.

Page 15: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 133

Large exophytic angiomyolipomas are anotherrelevant entity worthy of discussion. Angiomyoli-pomas, also known as renal hamartomas, are themost common benign mesenchymal tumors inthe kidney. These lesions contain mature adiposetissue, smooth muscle, and blood vessels. Occa-sionally, they can manifest as large exophytic renalmasses that contain varying degrees of fat and softtissue, mimicking the appearance of renal capsularliposarcoma. Given the different treatments andprognoses for these two lesions, distinguishing be-tween them radiologically is crucial. Because angio-myolipomas arise from the renal parenchymarather than the capsule, a defect in the parenchymais almost always visible at the site of the tumor’s or-igin (Fig. 25), whereas liposarcomas do not causesuch a defect. Angiomyolipomas also tend to bemore vascular than liposarcomas and are morelikely than liposarcoma to contain one or morelarge vessels within the lesion. Additional featuresthat suggest the diagnosis of angiomyolipoma in-stead of liposarcoma are the presence of intralesion-al hemorrhage, the smaller overall size of the lesion,and the presence of additional angiomyolipomaswithin the kidney [134].

Although rare, benign mesenchymal renal neo-plasms besides angiomyolipomas also can occur,including leiomyomas, lipomas, fibromas, neuro-genic tumors, and hemangiomas [122]. Renalleiomyomas, which most commonly occur in mid-dle-aged women, are the most common of thesetumors, and similar to their malignant counter-parts, they arise from smooth muscle cells located

Fig. 24. A 78-year-old man with high-grade spindlecell and pleomorphic sarcoma arising from the rightrenal capsule on pathology. Contrast-enhanced CTshows a peripherally located large right renal mass.Note the peritumoral vascularity (arrows) associatedwith this large mass.

in the renal capsule, pelvis, calyx, or blood vessels[135]; as a result, these tumors tend to be peripheralor parapelvic in location [136]. They have a variableradiographic pattern, which can range from an en-tirely solid to a purely cystic mass (Fig. 26) [136].Although leiomyomas are generally smaller andmore homogeneous than leiomyosarcomas, theirappearance can be similar [127,136]. Leiomyomasand leiomyosarcomas occasionally can contain

Fig. 25. A 63-year-old woman with right renal angio-myolipoma. Contrast-enhanced CT shows a large pre-dominantly fatty mass in the right kidney. Note thedefect in the renal parenchyma where the lesionarises from the kidney (arrow). This defect suggeststhe diagnosis of angiomyolipoma instead ofliposarcoma.

Fig. 26. A 49-year-old woman with incidental left re-nal leiomyoma arising from the left renal capsule onpathology. Contrast-enhanced CT image shows a ho-mogeneous mass (long arrow) arising peripherallyfrom the upper pole of the left kidney (short arrow).

Page 16: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al134

calcifications; these two entities cannot be confi-dently distinguished preoperatively unless gross in-vasion is observed, in which case the diagnosis ofleiomyoma is virtually eliminated [136].

Staging

Currently, the most commonly used staging systemis the TNM system [137] of the American JointCommittee on Cancer (Table 1). The patient’s over-all disease stage is determined by American JointCommittee on Cancer stage groupings (Table 2)[137].

Cancer-specific survival for patients treated withsurgery is highly correlated with the tumor stage(Table 3) [138]. Precise staging is crucial for preop-erative planning and prognosis. An overall stagingaccuracy of 91% by preoperative CT has been re-ported in the past, with most staging errors relatedto the diagnosis of perinephric extension of tumor[139]. Detection of perinephric tumor extensionhas no significant impact on therapy, but carriesprognostic significance. Currently, there is no reli-able indicator for perinephric tumor spread onCT. Perinephric stranding may be present in the ab-sence of tumor spread. One study showed that 50%of patients with tumor confined within the renalcapsule showed perinephric stranding [139]. Thepresence of an intact pseudocapsule, composed ofcompressed normal renal parenchyma and fibroustissue surrounding the renal mass, has been re-ported to be helpful in local staging of renal corticaltumors [140,141]. The pseudocapsule is best de-tected by T2-weighted imaging on MR imaging[140,141]. The presence of an intact pseudocapsulesuggests a lack of perinephric fat invasion [141].

CT has been found to have a high negative pre-dictive value for adrenal involvement from RCC[142]. It is suggested that only when the adrenalgland is not identified, not displaced or not en-larged on CT, adrenalectomy should be performedas part of radical nephrectomy, although even inthis select group adrenal involvement was presentin only 26% of the cases in one study [142].

Evaluation of the venous system in patients withrenal malignancy is crucial for treatment planning.Previously performed with conventional venogra-phy, this evaluation is now being done with nonin-vasive cross-sectional imaging. A thrombusinvolving the renal vein or inferior vena cava in a pa-tient with malignant renal tumor may represent tu-mor thrombus directly extending from the primarylocation or a bland blood clot or both. The presenceof enhancement within the thrombus indicates tu-mor thrombus, whereas bland thrombus wouldnot enhance after contrast administration (Figs.27 and 28). In some cases, the tumor thrombus

Table 1: TNM staging for renal cancer

T Primary tumor

TX Primary tumor cannotbe assessed

T0 No evidence of primarytumor

T1 Tumor %7 cm ingreatest dimension,limited to the kidney

T1a Tumor %4 cm ingreatest dimension,limited to the kidney

T1b Tumor >4 cm but %7 cmin greatest dimension,limited to the kidney

T2 Tumor >7 cm in greatestdimension, limited tothe kidney

T3 Tumor extends intomajor veins or invadesadrenal gland orperinephric tissues, butnot beyond Gerota’sfascia

T3a Tumor directly invadesadrenal gland orperirenal and/or renalsinus fat, but notbeyond Gerota’s fascia

T3b Tumor grossly extendsinto the renal vein or itssegmental (ie, muscle-containing) branches orthe vena cava below thediaphragm

T3c Tumor grossly extendsinto the vena cavaabove the diaphragm orinvades the wall of thevena cava

T4 Tumor invades beyondGerota’s fascia

N Regional lymph nodes

NX Regional lymph nodescannot be assessed

N0 No regional lymph nodemetastasis

N1 Metastasis in a singleregional lymph node

N2 Metastasis in >1regional lymph node

M Distant metastasis

MX Distant metastasiscannot be assessed

M0 No distant metastasisM1 Distant metastasis

Page 17: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 135

may extend beyond the margin of the inferior venacava, indicating invasion of the wall of the inferiorvena cava [143].

On cross-sectional imaging, assessment of lymphnodes relies on anatomic size criteria, and cross-sec-tional imaging is limited for detecting normal-sizedlymph nodes that harbor low-volume metastaticdisease or differentiating metastatic adenopathyfrom lymph node enlargement with a benign etiol-ogy. The sensitivity of CT for detection of regionallymph node metastases is reported to be 95%[144]. False-positive findings of 58% have been re-ported, however, when a size criterion of 1 cm isused for determining nodal metastasis, owing to re-active or other benign nodal changes [139,144].These false-positive findings were more frequentin patients with tumor involvement of the renalvein and tumor necrosis [144].

The most common metastatic sites from malig-nant renal cortical tumors include lung, bone,brain, liver, and mediastinum [145]. As a generalrule, small renal tumors are unlikely to present ini-tially with metastases, although there are occa-sional exceptions. CT of the abdomen and pelvisand a chest radiograph are essential studies inthe initial workup. A chest CT scan can be obtainedif the chest radiograph is abnormal, or advancedprimary disease is present. Despite advances inCT technology, the sensitivity for the detection ofpulmonary metastases from extrathoracic primary

Table 2: Stage grouping for renal cancer

Stage I T1, N0, M0Stage II T2, N0, M0Stage III T1, N1, M0

T2, N1, M0T3, N0, M0T3, N1, M0

Stage IV T4, N0, M0T4, N1, M0Any T, N2, M0Any T, any N, M1

Table 3: Survival rates for renal cancer by Tstage

T stage of renalmalignancy

Cancer-specificsurvival5-y 10-y

T1 95% 95%T2 88% 81%T3 59% 43%T4 20% 14%

tumors ranges from 75% to 95%, but is lower(50–70%) for pulmonary metastases smaller than6 mm [146,147]. In addition, pulmonary noduleswith benign causes may lead to false-positive re-sults. RCC is associated with hypervascular livermetastasis, particularly when the primary tumoris of the clear cell type. It has been shown that por-tal venous phase imaging detects 90% of livermetastases from RCC, with the addition of precon-trast or hepatic arterial phase imaging increasingthe sensitivity in lesion detection to almost 100%[148]. Rarely, advanced RCC also may metastasizeto the mesentery (Fig. 29). Bone metastases fromRCC are most commonly lytic (Fig. 30). Because85% of patients with bone metastases from RCCpresent with bone pain, a bone scan is not rou-tinely performed unless the patient complains ofbone pain or has an elevated serum alkaline phos-phatase. In addition, because osseous metastasesfrom RCC are usually lytic without osteoblasticactivity, bone scan may be negative in these cases.MR imaging may be considered in evaluation ofsymptomatic patients. Similarly, imaging of thebrain with CT or MR imaging is performed whenbrain metastasis is suggested by history or by phys-ical examination (Fig. 31).

The overall accuracy of MR imaging in staging iscomparable or superior to that of CT [71,95,149].Most likely because of its multiplanar capabilitiesand superior soft tissue contrast, MR imaging seemsto have advantages over CT in the evaluation of tu-mor vascular extension, the differentiation of peri-hilar lymph node metastases from vessels, and theassessment of direct tumor invasion into adjacentorgans [71,95,150]; MR imaging also can be usedto differentiate tumor reliably from bland throm-bus [150]. These capabilities make MR imaging anexcellent staging modality that should be usedwhen the CT findings are equivocal [71]. MR imag-ing is not accurate in indicating bowel and mesen-tery involvement, but technical advances and theintroduction of bowel contrast medium maychange this in the future [71].

Treatment planning

RCC is one of the few tumors in which well-docu-mented cases of spontaneous tumor regression inthe absence of therapy exist, and occasionally pa-tients with locally advanced or metastatic diseasemay exhibit indolent courses lasting several years,but this occurs very rarely and may not lead tolong-term survival. Surgical treatment is currentlythe only curative therapy for localized RCC andis indicated for patients with stage I, II, or IIIdisease.

Page 18: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al136

Fig. 28. MR images in a 77-year-old woman with a malignant left renal cortical tumor showing venous invasion.(A) Axial fat-suppressed T1-weighted image shows a left renal mass (short arrow) with tumor thrombus extend-ing into the left renal vein (long arrow). (B and C) Axial and coronal T2-weighted single-shot fast spin-echosequences show marked expansion of the inferior vena cava from tumor thrombus (arrows).

Fig. 27. A 78 year-old man with right clear cell renal carcinoma. (A) Contrast-enhanced CT shows a large hetero-geneous tumor (arrow) in the upper pole of the right kidney. (B) Contrast-enhanced CT at the level of the renalhilum shows a large expanding, enhancing tumor thrombus (arrow) in the right renal vein, bulging into theinferior vena cava.

Page 19: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 137

Fig. 29. A 67-year-old man with liver and peritoneal metastases from clear cell carcinoma. (A) Contrast-enhancedCT image shows multiple hepatic metastases. (B) Contrast-enhanced CT image shows a large heterogeneous met-astatic lesion to pancreatic tail (arrow). (C) Contrast-enhanced CT image shows peritoneal metastases (shortarrow).

Long-term survival can be achieved in about halfof patients with tumor extension into the vena cava,but it is thought that the prognosis of these patientsis primarily influenced by known adverse prognos-tic factors, such as capsular invasion, nodal disease,

and distant metastases, rather than vena caval tu-mor thrombus itself [138,151,152]. Imaging, espe-cially MR imaging, plays a crucial role indetermining the extent of caval thrombus becauseintracardiac extension requires the assistance of

Fig. 30. A 75-year-old man with osseous metastasis from clear cell renal carcinoma. (A) Unenhanced CT imageshows a lytic lesion in the left acetabulum. (B) Radiograph of left shoulder shows a lytic lesion in the proximalleft humerus with associated pathologic fracture.

Page 20: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al138

experienced cardiovascular surgeons for resectionand may entail venovenous or cardiopulmonarybypass techniques, with or without circulatory ar-rest [152]. Depending on the extent of tumorthrombus, perioperative mortality is high [152].

Because disease in the regional lymph nodes anddistant metastases have a great negative impact onsurvival, aggressive surgery in patients with grossnodal involvement or distant metastases is consid-ered unwarranted because it contributes little tosurvival [151]. Most patients with nodal involve-ment eventually relapse with distant metastases de-spite lymphadenectomy [153], but regional nodaldissection does provide diagnostic and prognosticinformation and the only potential curative ther-apy. In addition, enlarged lymph nodes on imagingcould have a benign etiology; patients with mild re-gional adenopathy on imaging still may be consid-ered for surgery [139,144]. In highly selected cases,surgical resection of locally recurrent RCC or a soli-tary metastasis may be associated with long-termsurvival [9,154].

Radical nephrectomy, defined as removal of allthe contents within the Gerota’s fascia, including

Fig. 31. A 67-year-old woman with brain metastasesfrom clear cell renal carcinoma. (A) Sagittal T1-weighted contrast-enhanced MR image of the brainshows an enhancing mass in the cerebrum. (B) AxialT1-weighted contrast-enhanced MR image of thebrain shows an enhancing mass in the cerebellum.

the kidney, perirenal fat, ipsilateral adrenal gland,and regional lymph nodes, has been the traditionalmainstay of surgical resection. More recent studieshave suggested, however, that in most patients adre-nal-sparing nephrectomy may be considered, withresection of the ipsilateral adrenal gland restrictedonly to patients with large upper-pole tumors or ab-normal-appearing adrenal glands on CT [142].

The long-term renal health of patients with renaltumors is an important clinical consideration forthe following reasons: (1) Renal cortical tumorsmay be bilateral and multifocal in hereditary andnonfamilial forms; (2) a significant number ofthese tumors carry a low metastatic potential andgood long-term prognosis [155]; and (3) as the pa-tients age, they are at risk for development of addedintrinsic renal damage from common diseases, suchas diabetes, hypertension, and glomerulonephritis[156]. Partial nephrectomy should be considereda diagnostic and therapeutic surgical approach forrenal cortical masses [32] whenever possible. Radi-cal nephrectomy for renal lesions that could beremoved by partial nephrectomy risk renal impair-ment in a substantial proportion of patients withbenign or relatively indolent disease. Although itused to be reserved for patients with bilateral tu-mors, renal insufficiency, or an anatomically orfunctionally solitary kidney, partial nephrectomyis now being offered as a standard surgical optionto all patients with renal lesions measuring 4 cmor smaller even in the setting of a normal contralat-eral kidney [157]. Evidence suggests that if techni-cally feasible, partial nephrectomy also should beconsidered for tumors 7 cm in size for better renalfunction preservation [158]. It has been shownthat disease-free survival rates are similar betweenrenal cortical tumors treated with radical and par-tial nephrectomy in properly selected patients[73], but the patients treated by partial nephrec-tomy have a decreased risk for chronic renal insuf-ficiency and proteinuria [73,156].

State-of-the-art multidetector CT provides thin-ner slices and allows for multiplanar three-dimen-sional image postprocessing, which may help inevaluating the anatomic relationship of the renalmass to adjacent structures, facilitating surgicalplanning [62,159,160]. This information is particu-larly crucial if nephron-sparing or laparoscopic sur-gery is being planned. State-of-the-art MR imagingalso has been shown to be accurate for the identifi-cation and characterization of renal neoplasmsamenable to partial nephrectomy [161].

Ultrasound also may assist in the preoperativeevaluation of renal cortical tumors. Renal ultrasoundis requested by the urologist as a template for theintraoperative ultrasound. It helps in selecting theproper surgical technique and in determining

Page 21: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 139

whether a partial nephrectomy can be attempted.Because of its multiplanar capability, ultrasoundcan show important landmarks and planes ofsections.

The prognosis of patients with renal cortical tu-mor depends on the mode of presentation (patientswith incidentally detected renal tumors have an ex-cellent prognosis), tumor size, histology, and stage[162,163]. The likelihood of renal tumor metasta-ses is probably associated with the size of the pri-mary tumor [164]. With improved imaging, renaltumors are being detected incidentally in greaternumbers and at smaller sizes and earlier stages[5–11]. Distant metastasis is unlikely in patientswith incidentally detected small renal masses. Inaddition, it seems that not all malignant renal tu-mors undergo significant active growth [6–8], andwithout significant tumor growth, the risk of metas-tasis may be limited as well.‘‘Watchful waiting’’ maybe a reasonable approach in highly selected cases[6,7], especially in patients who are elderly orpoor surgical candidates.

New imaging-guided therapeutic modalities,such as renal cryosurgery and radiofrequency abla-tion, are under active investigation [165]. It hasbeen reported that radiofrequency ablation cancompletely destroy renal cancers, while transmit-ting minimal collateral damage to surrounding re-nal parenchyma [165]. The preliminary results oftrials of imaging-guided radiofrequency thermal ab-lation or cryotherapy for treatment of primary renaltumors showed that these techniques may allowcomplete ablation of renal tumors with a low com-plication rate [165–167]. In addition, these studiesshowed that MR imaging is particularly useful formonitoring tumor destruction because of its excel-lent soft tissue contrast, high spatial resolution,multiplanar capabilities, high vascular conspicuity,and temperature sensitivity [166,167]. Untreatedareas can be accurately identified on MR imagingso that the electrode can be repositioned and addi-tional radiofrequency application performed in thesame treatment session, eliminating the need for re-turn visits for additional treatment of residual tu-mor, which are often required when ultrasound orCT is used for guidance [165–168]. Imaging-guidedlocal therapy also has been used successfully for thetreatment of local recurrences and isolated metasta-ses from RCC [165,169–171]. As with the treatmentof primary RCC, the long-term data on these imag-ing-guided therapeutic techniques is still limited,and questions remain regarding the optimal tech-niques and method of surveillance [172,173].These applications should be reserved for patientswho are poor surgical candidates and for whomstandard therapies have been exhausted[165,169,174].

The management of patients with advanced RCCremains a clinical challenge, with systemic therapyhaving only limited effectiveness [29]. There is noestablished role for adjuvant radiation or systemictherapy in patients treated with nephrectomy,even in patients with nodal involvement or incom-plete tumor resection [9,29,33,175]. No adjuvanttherapy has been shown to reduce the likelihoodof recurrence [176,177]. Radiation therapy with bi-sphosphonates may be considered for palliation ofpainful bone metastases.

Imaging follow-up

After surgical treatment, 20% to 30% of patientswith localized renal tumors relapse. Most recur-rences occur within 3 years after surgery, with themedian time to relapse being 1 to 2 years [178].Late tumor recurrence many years after initial treat-ment occasionally occurs. The lung is the most vul-nerable site for distant recurrence, which occurs in50% to 60% of patients (Fig. 32). Other commonsites of recurrence include bone, the nephrectomysite, brain, liver, and the contralateral kidney(Figs. 33–35) [179]. Greatest tumor diameter, Tstage, stage group, and nuclear grade are importantfactors in determining the likelihood of recurrence[179]. A long disease-free interval before recurrenceand metachronous presentation of recurrence arefavorable predictors of survival [154]. Efforts havebeen made to develop a nomogram to predict the5-year probability of freedom from recurrence forpatients with conventional clear cell RCC [180],which should be useful for patient counseling, clin-ical trial design, and effective patient follow-upstrategies.

As discussed previously, bilateral multifocal renaltumors are present in approximately 5% of patientswith sporadic renal tumors [48,49]. Although most

Fig. 32. A 70-year-old man with pulmonary metasta-ses 6 months after left nephrectomy for clear cellRCC. CT image shows multiple bilateral pulmonarynodules.

Page 22: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al140

Fig. 33. A 57-year-old man with recurrence 15 years after left nephrectomy for clear cell carcinoma. (A and B)Contrast-enhanced CT images show hypervascular hepatic (horizontal arrows), pancreatic (downward pointingvertical arrow) and right adrenal (upward pointing vertical arrow) metastases. Note surgical clips in left nephrec-tomy bed in (A).

bilateral tumors present synchronously, asynchro-nous lesions may occur many years after theoriginal nephrectomy, committing the patient tolong-term follow-up [25,48,155].

For imaging surveillance, CT is the modality ofchoice for detection of local recurrence and distantmetastases. In patients with compromised renalfunction or with contraindications to iodinatedcontrast, gadolinium-enhanced MR imaging of theabdomen and pelvis may be performed. A chestradiograph or chest CT study may be obtained forsurveillance of pulmonary metastases, based onpathologic stage and as clinically indicated.

FDG PET may have a potential role in the evalu-ation of distant metastases, especially when find-ings from conventional studies are equivocal, and

in the differentiation of recurrence from post-treat-ment changes [115,117,118,120]. Because of thehigh specificity and positive predictive value ofFDG PET, a positive result should be consideredstrongly suggestive of local recurrence or metastasis,although a negative result cannot reliably rule outmetastatic disease [120].

Summary

CT is the imaging modality of choice for evaluationof renal tumor. When a proper technique is used,CT provides high accuracy in detection of a renalmass. Certain imaging features and enhancementpatterns on CT may help distinguish different sub-types of renal tumors. In addition, CT provides

Fig. 34. A 55-year-old woman with local recurrence 3 years after right nephrectomy for clear cell carcinoma. (A)Axial T2-weighted single-shot fast spin-echo image shows a mass of mildly increased T2 signal extending fromright nephrectomy bed into right hepatic lobe (arrow). (B) Axial contrast-enhanced T1-weighted image with fatsaturation shows a heterogeneously enhancing mass extending from right nephrectomy bed into right hepaticlobe (arrow).

Page 23: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 141

useful diagnostic information for treatment plan-ning and follow-up. MR imaging and ultrasoundfunction as valuable problem-solving tools.

References

[1] American Cancer Society. Cancer facts and fig-ures 2006. Atlanta (GA): American Cancer Soci-ety; 2006.

[2] Ries LAG, Eisner MP, Kosary CL, et al. SEERcancer statistics review, 1975–2002. Bethesda(MD): National Cancer Institute; 2005.

[3] Provet J, Tessler A, Brown J, et al. Partial ne-phrectomy for renal cell carcinoma: indications,results and implications. J Urol 1991;145(3):472–6.

[4] Kosary CL, McLaughlin JK. Kidney and renalpelvis. In: Miller BA, Ries LAG, Hankey BG,et al, editors. SEER cancer statistics review,1973–1990. NIH publication no. 93-2789,XI.1-XI.22. Bethesda (MD): National CancerInstitute; 1993.

[5] Bosniak MA. Problems in the radiologic diagno-sis of renal parenchymal tumors. Urol ClinNorth Am 1993;20(2):217–30.

[6] Bosniak MA. Observation of small incidentallydetected renal masses. Semin Urol Oncol1995;13(4):267–72.

Fig. 35. A 59-year-old man with local recurrence 15months after right nephrectomy for clear cell carci-noma. (A) Axial T2-weighted single-shot fast spin-echo image shows a mass of intermediate T2 signalat the right nephrectomy bed (arrow). (B) Axial con-trast-enhanced T1-weighted image with fat satura-tion shows peripheral enhancement within thismass (arrow).

[7] Bosniak MA, Birnbaum BA, Krinsky GA, et al.Small renal parenchymal neoplasms: furtherobservations on growth. Radiology 1995;197(3):589–97.

[8] Kassouf W, Aprikian AG, Laplante M, et al. Nat-ural history of renal masses followed expec-tantly. J Urol 2004;171(1):111–3 [discussion113].

[9] Motzer RJ, Russo P, Nanus DM, Berg WJ. Renalcell carcinoma. Curr Probl Cancer 1997;21(4):185–232.

[10] Russo P. Renal cell carcinoma: presentation,staging, and surgical treatment. Semin Oncol2000;27(2):160–76.

[11] Smith SJ, Bosniak MA, Megibow AJ, et al. Renalcell carcinoma: earlier discovery and increaseddetection. Radiology 1989;170(3 Pt 1):699–703.

[12] Rabbani F, Reuter VE, Katz J, et al. Second pri-mary malignancies associated with renal cellcarcinoma: influence of histologic type. Urol-ogy 2000;56(3):399–403.

[13] Weiss LM, Gelb AB, Medeiros LJ. Adult renal ep-ithelial neoplasms. Am J Clin Pathol 1995;103(5):624–35.

[14] Kovacs G, Akhtar M, Beckwith BJ, et al. The Hei-delberg classification of renal cell tumours.J Pathol 1997;183(2):131–3.

[15] Russo P. Evolving understanding and surgicalmanagement of renal cortical tumors. MayoClin Proc 2000;75(12):1233–5.

[16] Russo P. Localized renal cell carcinoma. CurrTreat Options Oncol 2001;2(5):447–55.

[17] Lindblad P, Mellemgaard A, Schlehofer B, et al.International renal-cell cancer study: V. repro-ductive factors, gynecologic operations and ex-ogenous hormones. Int J Cancer 1995;61(2):192–8.

[18] Mandel JS, McLaughlin JK, Schlehofer B, et al.International renal-cell cancer study: IV. occu-pation. Int J Cancer 1995;61(5):601–5.

[19] McLaughlin JK, Chow WH, Mandel JS, et al. In-ternational renal-cell cancer study: VIII. role ofdiuretics, other anti-hypertensive medicationsand hypertension. Int J Cancer 1995;63(2):216–21.

[20] McLaughlin JK, Lindblad P, Mellemgaard A,et al. International renal-cell cancer study: I. to-bacco use. Int J Cancer 1995;60(2):194–8.

[21] Mellemgaard A, Lindblad P, Schlehofer B, et al.International renal-cell cancer study: III. role ofweight, height, physical activity, and use of am-phetamines. Int J Cancer 1995;60(3):350–4.

[22] Schlehofer B, Pommer W, Mellemgaard A, et al.International renal-cell-cancer study: VI. therole of medical and family history. Int J Cancer1996;66(6):723–6.

[23] Wolk A, Gridley G, Niwa S, et al. Internationalrenal cell cancer study: VII. Role of diet. Int JCancer 1996;65(1):67–73.

[24] Ishikawa I, Kovacs G. High incidence ofpapillary renal cell tumours in patients on

Page 24: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al142

chronic haemodialysis. Histopathology 1993;22(2):135–9.

[25] Rabbani F, Herr HW, Almahmeed T, et al. Tem-poral change in risk of metachronous contralat-eral renal cell carcinoma: influence of tumorcharacteristics and demographic factors. J ClinOncol 2002;20(9):2370–5.

[26] Lamiell JM, Salazar FG, Hsia YE. von Hippel-Lindau disease affecting 43 members of a singlekindred. Medicine (Baltimore) 1989;68(1):1–29.

[27] Cohen AJ, Li FP, Berg S, et al. Hereditary renal-cellcarcinoma associated with a chromosomal trans-location. N Engl J Med 1979;301(11):592–5.

[28] Pathak S, Strong LC, Ferrell RE, et al. Familialrenal cell carcinoma with a 3;11 chromosometranslocation limited to tumor cells. Science1982;217(4563):939–41.

[29] Motzer RJ, Bander NH, Nanus DM. Renal-cell carcinoma. N Engl J Med 1996;335(12):865–75.

[30] Beck SD, Patel MI, Snyder ME, et al. Effect ofpapillary and chromophobe cell type on dis-ease-free survival after nephrectomy for renalcell carcinoma. Ann Surg Oncol 2004;11(1):71–7.

[31] Eble JN, Bonsib SM. Extensively cystic renalneoplasms: cystic nephroma, cystic partially dif-ferentiated nephroblastoma, multilocular cysticrenal cell carcinoma, and cystic hamartoma ofrenal pelvis. Semin Diagn Pathol 1998;15(1):2–20.

[32] McKiernan J, Yossepowitch O, Kattan MW, et al.Partial nephrectomy for renal cortical tumors:pathologic findings and impact on outcome.Urology 2002;60(6):1003–9.

[33] Motzer RJ, Bacik J, Mariani T, et al. Treat-ment outcome and survival associated withmetastatic renal cell carcinoma of non-clear-cell histology. J Clin Oncol 2002;20(9):2376–81.

[34] Amin MB, Corless CL, Renshaw AA, et al.Papillary (chromophil) renal cell carcinoma:histomorphologic characteristics and evalua-tion of conventional pathologic prognosticparameters in 62 cases. Am J Surg Pathol 1997;21(6):621–35.

[35] Renshaw AA, Henske EP, Loughlin KR, et al.Aggressive variants of chromophobe renal cellcarcinoma. Cancer 1996;78(8):1756–61.

[36] Crotty TB, Farrow GM, Lieber MM. Chromo-phobe cell renal carcinoma: clinicopathologi-cal features of 50 cases. J Urol 1995;154(3):964–7.

[37] Kondagunta GV, Drucker B, Schwartz L, et al.Phase II trial of bortezomib for patients withadvanced renal cell carcinoma. J Clin Oncol2004;22(18):3720–5.

[38] Ficarra V, Righetti R, Pilloni S, et al. Prognosticfactors in patients with renal cell carcinoma:retrospective analysis of 675 cases. Eur Urol2002;41(2):190–8.

[39] Frank I, Blute ML, Cheville JC, et al. An out-come prediction model for patients with clearcell renal cell carcinoma treated with radical ne-phrectomy based on tumor stage, size, gradeand necrosis: the SSIGN score. J Urol 2002;168(6):2395–400.

[40] Gettman MT, Blute ML, Spotts B, et al. Patho-logic staging of renal cell carcinoma: signifi-cance of tumor classification with the 1997TNM staging system. Cancer 2001;91(2):354–61.

[41] Bretheau D, Lechevallier E, de Fromont M, et al.Prognostic value of nuclear grade of renal cellcarcinoma. Cancer 1995;76(12):2543–9.

[42] Ficarra V, Righetti R, Martignoni G, et al. Prog-nostic value of renal cell carcinoma nucleargrading: multivariate analysis of 333 cases.Urol Int 2001;67(2):130–4.

[43] Fiori E, De Cesare A, Galati G, et al. Prognosticsignificance of primary-tumor extension, stageand grade of nuclear differentiation in patientswith renal cell carcinoma. J Exp Clin Cancer Res2002;21(2):229–32.

[44] Fuhrman SA, Lasky LC, Limas C. Prognostic sig-nificance of morphologic parameters in renalcell carcinoma. Am J Surg Pathol 1982;6(7):655–63.

[45] Lohse CM, Blute ML, Zincke H, et al. Compar-ison of standardized and nonstandardized nu-clear grade of renal cell carcinoma to predictoutcome among 2,042 patients. Am J ClinPathol 2002;118(6):877–86.

[46] Ficarra V, Guille F, Schips L, et al. Proposal for re-vision of the TNM classification system for renalcell carcinoma. Cancer 2005;104(10):2116–23.

[47] Gudbjartsson T, Hardarson S, Petursdottir V,et al. Histological subtyping and nuclear grad-ing of renal cell carcinoma and their implica-tions for survival: a retrospective nation-widestudy of 629 patients. Eur Urol 2005;48:593–600.

[48] Patel MI, Simmons R, Kattan MW, et al. Long-term follow-up of bilateral sporadic renal tu-mors. Urology 2003;61(5):921–5.

[49] Richstone L, Scherr DS, Reuter VR, et al. Multi-focal renal cortical tumors: frequency, associ-ated clinicopathological features and impacton survival. J Urol 2004;171(2 Pt 1):615–20.

[50] Silver DA, Morash C, Brenner P, et al. Patho-logic findings at the time of nephrectomy forrenal mass. Ann Surg Oncol 1997;4(7):570–4.

[51] Dechet CB, Zincke H, Sebo TJ, et al. Prospectiveanalysis of computerized tomography and nee-dle biopsy with permanent sectioning to deter-mine the nature of solid renal masses inadults. J Urol 2003;169(1):71–4.

[52] Lechevallier E, Andre M, Barriol D, et al. Fine-needle percutaneous biopsy of renal masseswith helical CT guidance. Radiology 2000;216(2):506–10.

[53] Herts BR, Coll DM, Novick AC, et al. Enhance-ment characteristics of papillary renal

Page 25: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 143

neoplasms revealed on triphasic helical CT ofthe kidneys. AJR Am J Roentgenol 2002;178(2):367–72.

[54] Jinzaki M, Tanimoto A, Mukai M, et al. Double-phase helical CT of small renal parenchymalneoplasms: correlation with pathologic find-ings and tumor angiogenesis. J Comput AssistTomogr 2000;24(6):835–42.

[55] Kim JK, Kim TK, Ahn HJ, et al. Differentiationof subtypes of renal cell carcinoma on helicalCT scans. AJR Am J Roentgenol 2002;178(6):1499–506.

[56] Press GA, McClennan BL, Melson GL, et al. Pap-illary renal cell carcinoma: CT and sonographicevaluation. AJR Am J Roentgenol 1984;143(5):1005–9.

[57] Ruppert-Kohlmayr AJ, Uggowitzer M,Meissnitzer T, et al. Differentiation of renalclear cell carcinoma and renal papillary carci-noma using quantitative CT enhancement pa-rameters. AJR Am J Roentgenol 2004;183(5):1387–91.

[58] Sheir KZ, El-Azab M, Mosbah A, et al. Differen-tiation of renal cell carcinoma subtypes by mul-tislice computerized tomography. J Urol 2005;174(2):451–5 [discussion 455].

[59] Israel GM, Bosniak MA. Renal imaging for diag-nosis and staging of renal cell carcinoma. UrolClin North Am 2003;30(3):499–514.

[60] Silverman SG, Lee BY, Seltzer SE, et al. Small(< or 5 3 cm) renal masses: correlation of spiralCT features and pathologic findings. AJR Am JRoentgenol 1994;163(3):597–605.

[61] Kauczor HU, Schwickert HC, Schweden F, et al.Bolus-enhanced renal spiral CT: technique, di-agnostic value and drawbacks. Eur J Radiol1994;18(3):153–7.

[62] Sheth S, Scatarige JC, Horton KM, et al. Currentconcepts in the diagnosis and management ofrenal cell carcinoma: role of multidetector ctand three-dimensional CT. Radiographics2001;21(Spec No):S237–54.

[63] Schreyer HH, Uggowitzer MM, Ruppert-Kohlmayr A. Helical CT of the urinary organs.Eur Radiol 2002;12(3):575–91.

[64] Yuh BI, Cohan RH, Francis IR, et al. Compari-son of nephrographic with excretory phase heli-cal computed tomography for detecting andcharacterizing renal masses. Can Assoc RadiolJ 2000;51(3):170–6.

[65] Yuh BI, Cohan RH. Different phases of renalenhancement: role in detecting and characteriz-ing renal masses during helical CT. AJR Am JRoentgenol 1999;173(3):747–55.

[66] Yuh BI, Cohan RH. Helical CT for detection andcharacterization of renal masses. Semin Ultra-sound CT MR 1997;18(2):82–90.

[67] Szolar DH, Kammerhuber F, Altziebler S, et al.Multiphasic helical CT of the kidney: increasedconspicuity for detection and characterizationof small (<3-cm) renal masses. Radiology1997;202(1):211–7.

[68] Cohan RH, Sherman LS, Korobkin M, et al. Re-nal masses: assessment of corticomedullary-phase and nephrographic-phase CT scans. Radi-ology 1995;196(2):445–51.

[69] Birnbaum BA, Jacobs JE, Ramchandani P. Mul-tiphasic renal CT: comparison of renal mass en-hancement during the corticomedullary andnephrographic phases. Radiology 1996;200(3):753–8.

[70] Kopka L, Fischer U, Zoeller G, et al. Dual-phasehelical CT of the kidney: value of the corticome-dullary and nephrographic phase for evaluationof renal lesions and preoperative staging of re-nal cell carcinoma. AJR Am J Roentgenol1997;169(6):1573–8.

[71] Zhang J, Lefkowitz R, Ishill N, et al. Differentia-tion of solid renal cortical tumors by CT. Radi-ology, in press.

[72] Ambos MA, Bosniak MA, Valensi QJ, et al. An-giographic patterns in renal oncocytomas. Radi-ology 1978;129(3):615–22.

[73] Quinn MJ, Hartman DS, Friedman AC, et al. Re-nal oncocytoma: new observations. Radiology1984;153(1):49–53.

[74] Jinzaki M, Tanimoto A, Narimatsu Y, et al. An-giomyolipoma: imaging findings in lesionswith minimal fat. Radiology 1997;205(2):497–502.

[75] Daniel WW, Hartman GW, Witten DM, et al.Calcified renal mass: a review of ten years expe-rience at the Mayo Clinic. Radiology 1972;103:503–8.

[76] Bosniak MA. The current radiological approachto renal cysts. Radiology 1986;158(1):1–10.

[77] Brinker DA, Amin MB, de Peralta-Venturina M,et al. Extensively necrotic cystic renal cell carci-noma: a clinicopathologic study with compari-son to other cystic and necrotic renal cancers.Am J Surg Pathol 2000;24(7):988–95.

[78] Birnbaum BA, Maki DD, Chakraborty DP, et al.Renal cyst pseudoenhancement: evaluationwith an anthropomorphic body CT phantom.Radiology 2002;225(1):83–90.

[79] Maki DD, Birnbaum BA, Chakraborty DP, et al.Renal cyst pseudoenhancement: beam-harden-ing effects on CT numbers. Radiology 1999;213(2):468–72.

[80] Coulam CH, Sheafor DH, Leder RA, et al. Eval-uation of pseudoenhancement of renal cystsduring contrast-enhanced CT. AJR Am J Roent-genol 2000;174(2):493–8.

[81] Bae KT, Heiken JP, Siegel CL, et al. Renal cysts: isattenuation artifactually increased on contrast-enhanced CT images? Radiology 2000;216(3):792–6.

[82] Heneghan JP, Spielmann AL, Sheafor DH, et al.Pseudoenhancement of simple renal cysts:a comparison of single and multidetector heli-cal CT. J Comput Assist Tomogr 2002;26(1):90–4.

[83] Gokan T, Ohgiya Y, Munechika H, et al. Renalcyst pseudoenhancement with beam hardening

Page 26: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al144

effect on CT attenuation. Radiat Med 2002;20(4):187–90.

[84] Abdulla C, Kalra MK, Saini S, et al. Pseudoen-hancement of simulated renal cysts in a phan-tom using different multidetector CT scanners.AJR Am J Roentgenol 2002;179(6):1473–6.

[85] Pretorius ES, Wickstrom ML, Siegelman ES. MRimaging of renal neoplasms. Magn Reson Imag-ing Clin N Am 2000;8(4):813–36.

[86] Israel GM, Hindman N, Hecht E, et al. The useof opposed-phase chemical shift MRI in the di-agnosis of renal angiomyolipomas. AJR Am JRoentgenol 2005;184(6):1868–72.

[87] Zhang J, Israel GM, Krinsky GA, et al. Massesand pseudomasses of the kidney: imaging spec-trum on MR. J Comput Assist Tomogr 2004;28(5):588–95.

[88] Heiss SG, Shifrin RY, Sommer FG. Contrast-en-hanced three-dimensional fast spoiled gradient-echo renal MR imaging: evaluation of vascularand nonvascular disease. Radiographics 2000;20(5):1341–52 [discussion 1353–4].

[89] Semelka RC, Hricak H, Stevens SK, et al. Com-bined gadolinium-enhanced and fat-saturationMR imaging of renal masses. Radiology 1991;178(3):803–9.

[90] Eilenberg SS, Lee JK, Brown J, et al. Renalmasses: evaluation with gradient-echo Gd-DTPA-enhanced dynamic MR imaging. Radiol-ogy 1990;176(2):333–8.

[91] Semelka RC, Shoenut JP, Kroeker MA, et al. Re-nal lesions: controlled comparison between CTand 1.5-T MR imaging with nonenhanced andgadolinium-enhanced fat-suppressed spin-echoand breath-hold FLASH techniques. Radiology1992;182(2):425–30.

[92] Bosniak MA, Rofsky NM. Problems in the de-tection and characterization of small renalmasses. Radiology 1996;198(3):638–41.

[93] John G, Semelka RC, Burdeny DA, et al. Renalcell cancer: incidence of hemorrhage on MRimages in patients with chronic renal insuffi-ciency. J Magn Reson Imaging 1997;7(1):157–60.

[94] Scialpi M, Di Maggio A, Midiri M, et al. Smallrenal masses: assessment of lesion characteriza-tion and vascularity on dynamic contrast-enhanced MR imaging with fat suppression.AJR Am J Roentgenol 2000;175(3):751–7.

[95] Fein AB, Lee JK, Balfe DM, et al. Diagnosis andstaging of renal cell carcinoma: a comparison ofMR imaging and CT. AJR Am J Roentgenol1987;148(4):749–53.

[96] Outwater EK, Bhatia M, Siegelman ES, et al.Lipid in renal clear cell carcinoma: detectionon opposed-phase gradient-echo MR images.Radiology 1997;205(1):103–7.

[97] Outwater EK, Blasbalg R, Siegelman ES, et al.Detection of lipid in abdominal tissues withopposed-phase gradient-echo images at 1.5 T:techniques and diagnostic importance. Radio-graphics 1998;18(6):1465–80.

[98] Ho VB, Allen SF, Hood MN, et al. Renal masses:quantitative assessment of enhancement withdynamic MR imaging. Radiology 2002;224(3):695–700.

[99] Hecht EM, Israel GM, Krinsky GA, et al. Re-nal masses: quantitative analysis of enhance-ment with signal intensity measurementsversus qualitative analysis of enhancementwith image subtraction for diagnosing malig-nancy at MR imaging. Radiology 2004;232(2):373–8.

[100] Israel GM, Hindman N, Bosniak MA. Evalua-tion of cystic renal masses: comparison of CTand MR imaging by using the Bosniak classifica-tion system. Radiology 2004;231(2):365–71.

[101] Hartman DS, David CJ Jr, Goldman SM, et al.Renal lymphoma: radiologic-pathologic corre-lation of 21 cases. Radiology 1982;144(4):759–66.

[102] Forman HP, Middleton WD, Melson GL, et al.Hyperechoic renal cell carcinomas: increase indetection at US. Radiology 1993;188(2):431–4.

[103] Prasad SR, Saini S, Stewart S, et al. CT character-ization of ‘‘indeterminate’’ renal masses: tar-geted or comprehensive scanning? J ComputAssist Tomogr 2002;26(5):725–7.

[104] Helenon O, Chretien Y, Paraf F, et al. Renal cellcarcinoma containing fat: demonstration withCT. Radiology 1993;188(2):429–30.

[105] Helenon O, Merran S, Paraf F, et al. Unusual fat-containing tumors of the kidney: a diagnosticdilemma. Radiographics 1997;17(1):129–44.

[106] Strotzer M, Lehner KB, Becker K. Detection offat in a renal cell carcinoma mimicking angio-myolipoma. Radiology 1993;188(2):427–8.

[107] Taylor KJ, Ramos I, Carter D, et al. Correlationof Doppler US tumor signals with neovascularmorphologic features. Radiology 1988;166(1 Pt 1):57–62.

[108] Tamai H, Takiguchi Y, Oka M, et al. Contrast-enhanced ultrasonography in the diagnosis ofsolid renal tumors. J Ultrasound Med 2005;24(12):1635–40.

[109] Kim JK, Kim SH, Jang YJ, et al. Renal angiomyo-lipoma with minimal fat: differentiation fromother neoplasms at double-echo chemical shiftFLASH MR imaging. Radiology 2006;239(1):174–80.

[110] Raj GV, Bach A, Iasonos A, et al. Utility of pre-operative color Doppler ultrasonography in pre-dicting the histology of renal lesions. J Urol2007;177:53–8.

[111] Jinzaki M, Ohkuma K, Tanimoto A, et al. Smallsolid renal lesions: usefulness of power DopplerUS. Radiology 1998;209(2):543–50.

[112] Warshauer DM, McCarthy SM, Street L, et al.Detection of renal masses: sensitivities andspecificities of excretory urography/linear to-mography, US, and CT. Radiology 1988;169(2):363–5.

[113] Jamis-Dow CA, Choyke PL, Jennings SB, et al.Small (< or 5 3-cm) renal masses: detection

Page 27: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 145

with CT versus US and pathologic correlation.Radiology 1996;198(3):785–8.

[114] Malaeb BS, Martin DJ, Littooy FN, et al. Theutility of screening renal ultrasonography: iden-tifying renal cell carcinoma in an elderly asymp-tomatic population. BJU Int 2005;95(7):977–81.

[115] Aide N, Cappele O, Bottet P, et al. Efficiency of[(18)F]FDG PET in characterising renal cancerand detecting distant metastases: a comparisonwith CT. Eur J Nucl Med Mol Imaging 2003;30(9):1236–45.

[116] Goldberg MA, Mayo-Smith WW,Papanicolaou N, et al. FDG PET characteriza-tion of renal masses: preliminary experience.Clin Radiol 1997;52(7):510–5.

[117] Montravers F, Grahek D, Kerrou K, et al. Evalu-ation of FDG uptake by renal malignancies (pri-mary tumor or metastases) using a coincidencedetection gamma camera. J Nucl Med 2000;41(1):78–84.

[118] Ramdave S, Thomas GW, Berlangieri SU, et al.Clinical role of F-18 fluorodeoxyglucose posi-tron emission tomography for detection andmanagement of renal cell carcinoma. J Urol2001;166(3):825–30.

[119] Blake MA, McKernan M, Setty B, et al. Renal on-cocytoma displaying intense activity on 18F-FDG PET. AJR Am J Roentgenol 2006;186(1):269–70.

[120] Schoder H, Larson SM. Positron emission to-mography for prostate, bladder, and renal can-cer. Semin Nucl Med 2004;34(4):274–92.

[121] Caoili EM, Cohan RH, Inampudi P, et al. MDCTurography of upper tract urothelial neoplasms.AJR Am J Roentgenol 2005;184(6):1873–81.

[122] Rha SE, Byun JY, Jung SE, et al. The renal sinus:pathologic spectrum and multimodality imag-ing approach. Radiographics 2004;24(Suppl1):S117–31.

[123] Urban BA, Buckley J, Soyer P, et al. CT appear-ance of transitional cell carcinoma of the renalpelvis: Part 1. early-stage disease. AJR Am JRoentgenol 1997;169(1):157–61.

[124] Browne RF, Meehan CP, Colville J, et al. Transi-tional cell carcinoma of the upper urinary tract:spectrum of imaging findings. Radiographics2005;25(6):1609–27.

[125] Urban BA, Buckley J, Soyer P, et al. CT appear-ance of transitional cell carcinoma of the renalpelvis: Part 2. advanced-stage disease. AJR Am JRoentgenol 1997;169(1):163–8.

[126] Wagle DG, Moore RH, Murphy GP. Secondarycarcinomas of the kidney. J Urol 1975;114(1):30–2.

[127] Curry NS. Small renal masses (lesions smallerthan 3 cm): imaging evaluation and manage-ment. AJR Am J Roentgenol 1995;164(2):355–62.

[128] Israel GM, Krinsky GA. MR imaging of the kid-neys and adrenal glands. Radiol Clin North Am2003;41(1):145–59.

[129] Cohan RH, Dunnick NR, Leder RA, et al. Com-puted tomography of renal lymphoma. J Com-put Assist Tomogr 1990;14(6):933–8.

[130] Urban BA, Fishman EK. Renal lymphoma: CTpatterns with emphasis on helical CT. Radio-graphics 2000;20(1):197–212.

[131] Heiken JP, Gold RP, Schnur MJ, et al. Computedtomography of renal lymphoma with ultra-sound correlation. J Comput Assist Tomogr1983;7(2):245–50.

[132] Vogelzang NJ, Fremgen AM, Guinan PD, et al.Primary renal sarcoma in adults: a natural his-tory and management study by the AmericanCancer Society, Illinois Division. Cancer 1993;71(3):804–10.

[133] Shirkhoda A, Lewis E. Renal sarcoma and sarco-matoid renal cell carcinoma: CT and angio-graphic features. Radiology 1987;162(2):353–7.

[134] Israel GM, Bosniak MA, Slywotzky CM, et al.CT differentiation of large exophytic renal an-giomyolipomas and perirenal liposarcomas.AJR Am J Roentgenol 2002;179(3):769–73.

[135] Nagar AM, Raut AA, Narlawar RS, et al. Giantrenal capsular leiomyoma: study of two cases.Br J Radiol 2004;77(923):957–8.

[136] Steiner M, Quinlan D, Goldman SM, et al. Leio-myoma of the kidney: presentation of 4 newcases and the role of computerized tomogra-phy. J Urol 1990;143(5):994–8.

[137] Kidney. In: American Joint Committee on Can-cer. AJCC Cancer Staging Manual. 6th edition.New York: Springer; 2002. p. 323–5.

[138] Javidan J, Stricker HJ, Tamboli P, et al. Prognos-tic significance of the 1997 TNM classificationof renal cell carcinoma. J Urol 1999;162(4):1277–81.

[139] Johnson CD, Dunnick NR, Cohan RH, et al. Re-nal adenocarcinoma: CT staging of 100 tumors.AJR Am J Roentgenol 1987;148(1):59–63.

[140] Yamashita Y, Honda S, Nishiharu T, et al. De-tection of pseudocapsule of renal cell carci-noma with MR imaging and CT. AJR Am JRoentgenol 1996;166(5):1151–5.

[141] Roy C Sr, El Ghali S, Buy X, et al. Significance ofthe pseudocapsule on MRI of renal neoplasmsand its potential application for local staging:a retrospective study. AJR Am J Roentgenol2005;184(1):113–20.

[142] Gill IS, McClennan BL, Kerbl K, et al. Adrenalinvolvement from renal cell carcinoma: predic-tive value of computerized tomography. J Urol1994;152(4):1082–5.

[143] Didier D, Racle A, Etievent JP, et al. Tumorthrombus of the inferior vena cava secondaryto malignant abdominal neoplasms: US andCT evaluation. Radiology 1987;162(1 Pt 1):83–9.

[144] Studer UE, Scherz S, Scheidegger J, et al. En-largement of regional lymph nodes in renalcell carcinoma is often not due to metastases.J Urol 1990;144(2 Pt 1):243–5.

Page 28: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Zhang et al146

[145] Hilton S. Imaging of renal cell carcinoma.Semin Oncol 2000;27(2):150–9.

[146] Diederich S, Semik M, Lentschig MG, et al. He-lical CT of pulmonary nodules in patients withextrathoracic malignancy: CT-surgical correla-tion. AJR Am J Roentgenol 1999;172(2):353–60.

[147] Margaritora S, Porziella V, D’Andrilli A, et al.Pulmonary metastases: can accurate radiologi-cal evaluation avoid thoracotomic approach?Eur J Cardiothorac Surg 2002;21(6):1111–4.

[148] Raptopoulos VD, Blake SP, Weisinger K, et al.Multiphase contrast-enhanced helical CT ofliver metastases from renal cell carcinoma.Eur Radiol 2001;11(12):2504–9.

[149] Semelka RC, Shoenut JP, Magro CM, et al.Renal cancer staging: comparison of contrast-enhanced CT and gadolinium-enhanced fat-suppressed spin-echo and gradient-echo MRimaging. J Magn Reson Imaging 1993;3(4):597–602.

[150] Myneni L, Hricak H, Carroll PR. Magnetic reso-nance imaging of renal carcinoma with exten-sion into the vena cava: staging accuracy andrecent advances. Br J Urol 1991;68(6):571–8.

[151] Cherrie RJ, Goldman DG, Lindner A, et al.Prognostic implications of vena caval extensionof renal cell carcinoma. J Urol 1982;128(5):910–2.

[152] Bissada NK, Yakout HH, Babanouri A, et al.Long-term experience with management of re-nal cell carcinoma involving the inferior venacava. Urology 2003;61(1):89–92.

[153] Phillips E, Messing EM. Role of lymphadenec-tomy in the treatment of renal cell carcinoma.Urology 1993;41(1):9–15.

[154] Kavolius JP, Mastorakos DP, Pavlovich C, et al.Resection of metastatic renal cell carcinoma. JClin Oncol 1998;16(6):2261–6.

[155] Grimaldi G, Reuter V, Russo P. Bilateral non-familial renal cell carcinoma. Ann Surg Oncol1998;5(6):548–52.

[156] McKiernan J, Simmons R, Katz J, et al. Naturalhistory of chronic renal insufficiency after par-tial and radical nephrectomy. Urology 2002;59(6):816–20.

[157] Gilbert SM, Russo P, Benson MC, et al. Theevolving role of partial nephrectomy in themanagement of renal cell carcinoma. Curr On-col Rep 2003;5(3):239–44.

[158] Dash A, Vickers AJ, Schachter LR, et al. Com-parison of outcomes in elective partial vs radi-cal nephrectomy for clear cell renal cellcarcinoma of 4–7 cm. BJU Int 2006;97(5):939–45.

[159] Chernoff DM, Silverman SG, Kikinis R, et al.Three-dimensional imaging and display of re-nal tumors using spiral CT: a potential aid topartial nephrectomy. Urology 1994;43(1):125–9.

[160] Lee CT, Hilton S, Russo P. Renal mass withina horseshoe kidney: preoperative evaluation

with three-dimensional helical computedtomography. Urology 2001;57(1):168.

[161] Pretorius ES, Siegelman ES, Ramchandani P,et al. Renal neoplasms amenable to partial ne-phrectomy: MR imaging. Radiology 1999;212(1):28–34.

[162] Lee CT, Katz J, Fearn PA, et al. Mode of presen-tation of renal cell carcinoma provides prog-nostic information. Urol Oncol 2002;7(4):135–40.

[163] Kattan MW, Reuter V, Motzer RJ, et al. A postop-erative prognostic nomogram for renal cell car-cinoma. J Urol 2001;166(1):63–7.

[164] Duffey BG, Choyke PL, Glenn G, et al. The re-lationship between renal tumor size and metas-tases in patients with von Hippel-Lindaudisease. J Urol 2004;172(1):63–5.

[165] Zagoria RJ, Hawkins AD, Clark PE, et al. Percu-taneous CT-guided radiofrequency ablation ofrenal neoplasms: factors influencing success.AJR Am J Roentgenol 2004;183(1):201–7.

[166] Lewin JS, Nour SG, Connell CF, et al. Phase IIclinical trial of interactive MR imaging-guidedinterstitial radiofrequency thermal ablation ofprimary kidney tumors: initial experience. Radi-ology 2004;232(3):835–45.

[167] Silverman SG, Tuncali K, vanSonnenberg E,et al. Renal tumors: MR imaging-guided percu-taneous cryotherapy—initial experience in 23patients. Radiology 2005;236(2):716–24.

[168] Gervais DA, McGovern FJ, Wood BJ, et al. Ra-dio-frequency ablation of renal cell carcinoma:early clinical experience. Radiology 2000;217(3):665–72.

[169] Zagoria RJ. Percutaneous image-guided radio-frequency ablation of renal malignancies. Ra-diol Clin North Am 2003;41(5):1067–75.

[170] McLaughlin CA, Chen MY, Torti FM, et al. Ra-diofrequency ablation of isolated local recur-rence of renal cell carcinoma after radicalnephrectomy. AJR Am J Roentgenol 2003;181(1):93–4.

[171] Zagoria RJ, Chen MY, Kavanagh PV, et al. Radiofrequency ablation of lung metastases from re-nal cell carcinoma. J Urol 2001;166(5):1827–8.

[172] Wagner AA, Solomon SB, Su LM. Treatment ofrenal tumors with radiofrequency ablation.J Endourol 2005;19(6):643–52 [discussion652–3].

[173] Varkarakis IM, Allaf ME, Inagaki T, et al. Percu-taneous radio frequency ablation of renalmasses: results at a 2-year mean followup.J Urol 2005;174(2):456–60 [discussion 460].

[174] Su LM, Jarrett TW, Chan DY, et al. Percutaneouscomputed tomography-guided radiofrequencyablation of renal masses in high surgical riskpatients: preliminary results. Urology 2003;61(4, Suppl 1):26–33.

[175] Motzer RJ, Russo P. Systemic therapy for renalcell carcinoma. J Urol 2000;163(2):408–17.

[176] Messing EM, Manola J, Wilding G, et al. PhaseIII study of interferon alfa-NL as adjuvant

Page 29: 06 - Radiol Clin N Am 2007 - Imaging of Kidney Cancer

Imaging of Kidney Cancer 147

treatment for resectable renal cell carcinoma:an Eastern Cooperative Oncology Group/Inter-group trial. J Clin Oncol 2003;21(7):1214–22.

[177] Clark JI, Atkins MB, Urba WJ, et al. Adjuvanthigh-dose bolus interleukin-2 for patientswith high-risk renal cell carcinoma: a cytokineworking group randomized trial. J Clin Oncol2003;21(16):3133–40.

[178] Sandock DS, Seftel AD, Resnick MI. A new pro-tocol for the followup of renal cell carcinoma

based on pathological stage. J Urol 1995;154(1):28–31.

[179] Chae EJ, Kim JK, Kim SH, et al. Renal cellcarcinoma: analysis of postoperative recur-rence patterns. Radiology 2005;234(1):189–96.

[180] Sorbellini M, Kattan MW, Snyder ME, et al. Apostoperative prognostic nomogram predictingrecurrence for patients with conventional clearcell renal cell carcinoma. J Urol 2005;173(1):48–51.