Advances in Renal Imaging 2003 PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 207

Radiol Clin N Am 41 (2003) xi – xii

Preface
Advances in renal imaging

Philip J. Kenney, MD
Guest Editor

Upon being invited to edit an issue of the Radio- there is little debate about which study to perform. In
logic Clinics of North America on renal imaging, it some other areas, the authors properly include more
was my intention to produce a work that not only discussion on the advantages and disadvantages of
presented the current state-of-the-art but also gave a different methods, commonly CT versus MR imaging,
glimpse of the future. With contributions from excel- as well as important discussion as to who should be
lent clinician/scientist radiologists, I believe those evaluated. These relate particularly to evaluation of
goals have been met. hypertension in the adult by Drs. Hartman, Kawa-
It is striking how much of this issue is devoted to shima, and King and in the pediatric population by
CT and MR imaging in various forms, with some Drs. Roth, Spottswood, Chan, and Roth, and the
ultrasound. Technical advances, particularly multi- evaluation of hematuria included in the presentation
detector row CT, have had a major impact on the eval- of CT urography and MR urography by Drs. Kawa-
uation of renal disorders. Today, CT and MR imaging shima, Glockner, and King.
have, to a great degree, replaced ‘‘standard’’ intra- While in truth all of the contributors offer some
venous urography. Several of the sections in this issue glimpse of the future, many of the sections concen-
are technically oriented, especially those from Drs. trate on the techniques whereby state-of-the-art cross-
Lockhart and Smith, Drs. Zhang, Pedrosa, and Rofsky, sectional imaging methods can replace and surpass
and Drs. Huang and Lee, as well as Drs. Kawashima, the radiographic technology (intravenous urography
Glockner, and King and Drs. Hartman, Kawashima, and catheter angiography) of the past. However, the
and King. Technical developments in CT and MR discussions of MR imaging of renal function by
imaging now allow for excellent diagnostic capabili- Drs. Huang and Lee, and renal imaging with ultra-
ties for renal disorders, but detailed specifics of the sound contrast present techniques whereby previously
technique related to the disorder being sought must be available technology can be used to derive new infor-
understood to attain high accuracy. mation. Combination of anatomic and functional
Two sections, those on renal trauma by Dr. Smith information can lead to new uses of imaging.
and urinary lithiasis by Dr. Kenney, could be consid- Finally, although many of the sections deal with di-
ered state-of-the-art presentations in which CT has agnosis of the renal mass (and properly so, considering
clearly demonstrated its primacy, now considered the this is a common problem), several sections expand the
‘‘one and only’’ in these circumstances. Although perspective. It is not enough for the radiologist today to
some controversies remain in these areas, and under- understand technique and interpretation for accurate
standing of correct technique remains important, diagnosis; one must also have some understanding of

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00122-2
xii P.J. Kenney / Radiol Clin N Am 41 (2003) xi–xii

the basic principles of the disease, including genetics, Philip J. Kenney, MD


as well as the treatment options. In contributions from Department of Diagnostic Radiology
Drs. Choyke, Zagoria, and El-Galley, a deeper under- University of Alabama at Birmingham
standing is provided of the various diseases called JT N370
renal carcinoma, and of the wide variety of treatment 619 19th Street South
options now available, whether provided directly by a Birmingham, AL 35249-6830, USA
radiologist or by urologic surgeons. E-mail address: pkenney@uabmc.edu
Radiol Clin N Am 41 (2003) 863 – 875

Technical considerations in renal CT


Mark E. Lockhart, MD, MPH*, J. Kevin Smith, PhD, MD
Department of Radiology, University of Alabama at Birmingham, 619 19th Street, South JTN363, Birmingham,
AL 35249 – 6830, USA

The use of radiologic imaging, specifically CT, processor capability, and faster and larger data storage
continues to grow in diagnostic importance. The methods have also been developed. Reconstruction
impact of CT on medicine and urologic evaluation times have approached real-time review of the data.
has transformed it from an exotic tool into a diag- More recently developed scanners use multiple rows
nostic cornerstone. Few patients with urinary symp- of detectors to capture multiple image slices from
toms or signs escape diagnostic imaging, and most the single beam of radiation that passes through a
undergo multiple examinations. For decades, intra- patient. The entire abdomen can be studied in less than
venous (IV) pyelogram was the primary means of 15 seconds, within a single breathhold. With increased
noninvasive evaluation of the upper urologic system. image speed and quality, CT has made major inroads
In the last 20 years, however, the application of CT into vascular imaging. New developments include
in urology has exploded. In many institutions, CT plate detector technology, which allows volumetric
has largely replaced the use of IV pyelogram. This reconstruction in any plane, a long-time advantage of
article reviews and discusses technical considera- MR imaging over CT.
tions in the performance of CT for various urologic
clinical indications.
CT has undergone many changes and improve- Basic concepts
ments since its development. The earliest scanners
used axial imaging and a single beam of radiation and Pitch
required minutes to reconstruct the most basic
images. As the scanners were improved, the speed Pitch is a term that is used to describe the relative
of acquisition and image quality grew proportionally. movement of the patient as the x-ray source and
The medical benefits and number of uses became detector circle to acquire data. For axial CT, the pitch
widely accepted, and by the late 1990s CT was firmly is zero because the table is not moving as the data
entrenched as a primary diagnostic tool. One major are acquired. Single-detector helical CT pitch is deter-
improvement was the use of slip-ring technology to mined by table speed in centimeters per second
create helical data acquisition as the patient was divided by slice thickness.
slowly moved through the rotating radiation beam. The calculation of pitch in multidetector is more
Subsecond scanners were developed and when com- complicated and different manufacturers use two
bined with slip ring helical scanning was fast enough separate methods of description. On General Electric
to follow contrast boluses through the vessels for CT scanners (GE Medical Systems, Milwaukee, WI), for
angiography (CTA). Today, thinner slices require example, the pitch is expressed as the table speed
additional radiation and yield more images than divided by the nominal slice thickness. On current
ever. Larger heat capacity tubes, increased computer four-channel CTs, however, the total x-ray beam
width is four times the nominal slice width. For
example, a 1.25-mm slice thickness study with a
* Corresponding author. table speed of 7.5 mm per gantry rotation yields a
E-mail address: mlockhar@uabmc.edu (M.E. Lockhart). pitch of 6, 7.5 mm per 1.25 mm. A more widely

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00065-4
864 M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875

accepted definition (eg, used on Siemens [Erlangen, the part of the x-ray beam that strikes the outside
Germany] CT scanners) of pitch is the table speed per detectors passes through the patient at more of an
rotation divided by the x-ray beam width. For a four- angle than the part of the x-ray beam that strikes the
channel system with 1.25-mm slice thickness this central detectors, the so-called ‘‘cone beam’’ effect.
technique has a beam width of 4  1.25 mm = 5 This angle leads to errors in image reconstruction and
mm, and this definition yields a pitch of 1.5, or 7.5 mm artifacts if not taken into account, so multislice
per 5 mm. For subsecond scanners the length of scanners must either scan at certain pitches that
coverage increases relative to a 1-second scanner cancel out these effects or use special cone beam
proportionally for the same technique. A 0.5-second image reconstruction algorithms.
scanner can cover twice the distance in the same time The raw helical CT data are generally recon-
as a 1-second CT using similar technique. structed into axial images and these images may then
An equivalent to 1 pitch for a single-detector CT be used to generate images in any plane, but the off-
should be used in the evaluation or characterization of axis images are often suboptimal if the original slices
renal lesions to decrease volume averaging from slice are too thick. With thinner slices and high milli-
profile broadening. A pitch 1.5 to 2 is often used to amperes and kilovolt (peak), high-quality reconstruc-
cover rapidly an adequate volume for CT angiogra- tions are possible. New workstations and software
phy or trauma evaluation. This allows increased have greatly reduced reconstruction time, especially
coverage speed to track the contrast bolus without in vascular imaging.
much detriment to effective slice thickness [1]. The volumetric acquisition is beneficial in opti-
mizing of even axially oriented images with regard to
Detectors a specific lesion. Thinner slice reconstructions may
be adjusted to center the slice volume on the lesion to
There are now a large variety of detector configu- reduce volume averaging with the adjacent paren-
rations in use. Currently, new scanners generally use chyma and provide more accurate assessment of
a rotating radiation source with an attached arc of lesion enhancement. This may be performed retro-
rotating detectors that are composed of multiple spectively on the data set without the resultant
contiguous detector rows. There is wide variation in increased radiation of rescanning the patient.
the number of detector arrays produced by various
manufacturers. Some scanners have equally sized
detectors and others have smaller detectors in the
central beam region with larger detectors at the Contrast selection and types
periphery. Each of the systems allows the combina-
tion of several detectors to allow images of different Routine protocols: concentration and total dosage
slice thickness during image acquisition. In addition,
there is variation in the number of helices generated In routine applications, approximately 125 to
based on the number of rows of detectors and 150 mL iodinated contrast is given by reliable IV
separate data channels. The number of helices has access. At a standard 2 to 3 mL/second injection rate,
increased from 1 to 2 (dual-slice) to 4 and now 16 in the contrast bolus lasts 60 to 75 seconds. Both high-
the last decade. There will soon be availability of and low-osmolar IV contrast result in satisfactory
32-row CT scanners with developments of still more image quality. Initially, low-osmolar contrast was
rows of detectors and channels to allow even faster very expensive, and selective usage of low-osmolar
imaging in the near future. contrast was the norm because of the large cost
difference between high- and low-osmolar contrast
Reconstruction agents. Patients with increased risk of allergy, renal
insufficiency, or other indications were given low
In single-detector helical CT or multidetector-row osmolar, whereas the standard was to give high-
CT (MDCT), the raw data are acquired at a different osmolar contrast. As the price difference has dimin-
level of the patient from each vantage point depend- ished, many institutions have selected low-osmolar
ing on where the x-ray source is located in its circle. agents for all of their routine contrasted CTs.
To calculate a slice of image data that seems axial For patients with decreased renal function, lower
rather than a helix, the data from different points in contrast volumes are often given to reduce the risk of
the helix are interpolated to the displayed level. This contrast-induced nephrotoxicity. There is no degra-
interpolation leads to slice profile broadening and dation of enhancement when the contrast dosage is
may also lead to artifacts. With multislice scanners 1.5 mL/kg or greater [2], and with faster multislice
M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875 865

scanners it may be possible further to reduce the dose injected if an extravasation of contrast outside the
for such studies as CTA. vein occurs.

Injection rates Oral contrast versus water

The current standard of care for abdominal CT In renal imaging, the choice of oral contrast agent
uses controlled IV bolus of contrast by a power is less critical than in gastrointestinal imaging. There
injector. For routine imaging of nonspecific renal are cases, however, where the choice of oral contrast
complaints or characterization of a renal mass, the can impact a renal CT. In evaluation of renal calculi,
standard infusion rate is 2 to 3 mL/second. This can dense oral contrast in the small bowel can make
be accomplished through any number of IV access detection of a ureteral calculus more difficult. CTA
sites. Injection through a small (3 to 4F catheter is another clinical situation where positive oral con-
diameter) peripherally inserted central catheter is trast can be counterproductive; no oral contrast is
not practical because of the small bore and length given, or water may be selected. The density of oral
of the catheter and the viscosity of the contrast [3]. contrast may lie adjacent to vessels and significantly
The injection rate may also be limited for some other increases the difficulty of three-dimensional recon-
central venous catheters. Concern for power injection struction (Fig. 2). For renal donor evaluation and
of contrast directly into the cardiac chambers is renal mass evaluation of vascular supply for surgical
present because of the possibility of inducing a planning, water is preferable to positive oral contrast
cardiac arrhythmia, but studies have suggested that for bowel distention.
central catheter injection of contrast is safe with
adequate injection rate guidelines [4,5]. High-concentration contrast: new uses
If CTA is needed, higher injection rates are
necessary to opacify the arteries densely. Typically, Recent research has used high-concentration con-
a rate of 3 to 5 mL/second is selected for CTA imaging trast agent for CT imaging. The most obvious appli-
of the renal vessels (Fig. 1). This requires a reliable cation is for CTA. Dense agents can be used to allow
20-gauge or larger IV line in a good antecubital vein. improved opacification of small vessels to allow
An 18-gauge IV line can allow up to 7 mL/second, but reconstruction of the vascular anatomy. In the urologic
the authors do not use this rapid rate on their CTA system, this may allow demonstration of vascular
studies. Before power injection, their technologists abnormalities, such as arteriovenous malformations;
test the IV line with a rapid hand injection of saline to small aneurysms; tiny accessory arteries; and subtle
evaluate for any extravasation. Pressure limits are vascular abnormalities, such as mild fibromuscular
used for the power injection to limit the volume dysplasia. Other possible applications include demon-

Fig. 1. CT and CT angiography at the level of the kidneys demonstrate differences in enhancement using routine and high-rate
injection rates. (A) Standard injection rate of contrast 2 to 3 mL/second with good contrast enhancement of the vessels and renal
parenchyma. (B) High-rate injection of contrast 4 to 5 mL/second with arterial timing using same CT scanner in the same patient
at a different date demonstrates higher density within the aorta and renal vessels. Denser cortical enhancement also is noted.
866 M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875

stration of artery course and appearance to help


characterize renal masses.

CTA: technical differences

CT angiography optimizes CT technique for the


opacification and display of the arterial system. Tech-
nical differences include timing of the contrast bolus,
slice thickness, dose, pitch, and reconstructions.
Two methods are generally used for selecting the
optimal timing for CTA imaging: test injection or
bolus tracking. For test injection, a small-volume
bolus is injected at a similar rate to the desired
CTA injection rate. A single level low-dose CT scan
Fig. 2. Reconstruction images in the coronal plane of CT
angiography in a patient who received iodinated oral contrast is repeatedly imaged through the aorta at the area of
before the study. The overlying dense contrast within the interest. This technique generally uses less than
bowel increases the difficulty to visualize the abdominal 100 kV and less than 100 mA performed every 2 to
vessels separately. 3 seconds [6]. Once peak enhancement is noted, the
calculated interval is selected as the delay for imag-
ing. Alternatively, bolus tracking does not use a test
injection; rather, the diagnostic bolus is tracked using
similar low-dose repetitive imaging. When contrast
reaches the left ventricle or proximal aorta depending

Fig. 3. Reconstruction techniques at the level of the kidney with renal artery aneurysm for CT angiography using (A) maximum
intensity projection, (B) shaded surface reconstruction, and (C) volume rendering.
M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875 867

on area of interest, the scan is initiated either auto- healthy patients, CTA can characterize the arterial and
matically or manually. The tracking level is generally venous supply to the kidneys to help select the easiest
chosen upstream from the area of interest because and safest kidney for renal donation.
there is a slight delay before images are obtained as
the scanner moves to the starting position and
changes anode current. Helical images then ‘‘chase’’ Timing and technique issues
the contrast bolus as it flows farther from the heart.
At each level the arteries are densely opacified during Nonenhanced CT for urolithiasis
the time of imaging. Thin slices (usually 1 to 3 mm
thick) are obtained with sufficient radiographic tech- Noncontrast CT has become the primary radio-
nique such that mottle is not limiting. In the standard logic study for the evaluation of renal stones. Some
helical CT and some early MDCT, a higher pitch may specific technical issues remain unresolved. In the
be necessary to keep up with the contrast and to scan evaluation of known renal calculi with renal colic or
the area of interest within the heat capacity of the hematuria, CT without IV or oral contrast is usually
x-ray tube and the breathhold capacity of the patient. adequate and quick. In a significant proportion of
With newer scanners, there is the possibility of out- patients with less specific presentation, no stone is
running a tight contrast bolus if the scan is too fast or found. Many of these patients may eventually receive
started too early after the bolus arrival. a CT with oral and IV contrast. If no oral contrast was
Oral contrast is not given in CTA so that the only given for the initial renal stone CT, the patient must
dense contrast is within the arteries. This is essential be moved from the scanner and given time to drink
for ease of three-dimensional reconstruction, which the oral contrast to optimize the contrasted study. The
uses threshold selection, maximum intensity projec- administration of oral contrast before renal stone
tion, or volume rendering for reconstructions. If protocol may allow the IV contrasted study imme-
necessary to answer additional clinical questions, diately to follow the stone study, reducing patient wait
water can be given as an oral agent. Although and improving study time efficiency. Oral contrast,
multiple phases of imaging can be performed using however, has the potential to mask a ureteral calculus
high-capacity heat tubes, the thin slices and high that abuts bowel.
radiographic technique may result in substantial dose Occasionally a calcification is noted in the pelvis,
to the patient. but the ureters cannot be followed at the level of the
A limitation of traditional CTA is the suboptimal calcification. In these cases, limited images of the
timing of solid organ enhancement. The technique is pelvis may be performed at 3 to 5 minutes after low-
less sensitive for renal masses compared with nephro- dose IV contrast to identify the ureters and determine
graphic phase imaging. Central masses especially whether the calcification is vascular or urologic. If the
may be missed because the hypoenhancing mass symptoms are less typical of renal colic, it may be
may be indistinguishable from the relatively hypo- preferable to include the upper abdomen on con-
enhancing renal medulla. trasted imaging to help exclude other possible causes
Reconstructions are often performed to present of abdominal symptoms. In one series, 13.1% of
images that resemble angiography. Reconstruction unenhanced CT for evaluation of urinary tract calculi
techniques that are commonly used are maximum subsequently received IV contrast [7].
intensity projection, shaded surface reconstruction, Another question arises in the setting of hematuria
and volume rendering (Fig. 3). At the authors’ institu- without definite renal colic. In these patients, a non-
tion, a trained technologist rapidly reconstructs the contrast renal stone CT may be a reasonable first
images. New workstations have streamlined the pro- study. The absence of renal calculi, however, should
cess, and are much faster and more user friendly than prompt a CT with IV contrast. In the study by Gottlieb
even systems from a few years ago. Life-like three- et al [7], approximately 6% of studies for urinary
dimensional images may be constructed from various calculi demonstrated a nonurologic cause for symp-
perspectives including a surgical viewpoint for the toms, such as appendicitis or diverticulitis. The timing
specific procedure in question. Images showing rota- is usually at 70 to 90 seconds after injection to exclude
tion of the three-dimensional volume or progressive renal tumor or infection as an etiology. Pelvis images
removal of overlying structures may be helpful. The are performed after a 3-minute delay to allow filling of
reconstructions can be very useful in localizing a renal the urinary bladder with contrast.
tumor for segmental or laparoscopic resection. The If the collecting systems have fluid-filled struc-
relationships of the mass to the adjacent structures, tures, and parapelvic cysts versus hydronephrosis are
such as chest wall, can be demonstrated exquisitely. In a consideration, a postcontrast scanogram scout can
868 M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875

Fig. 4. Corticomedullary phase CT of kidney (A) shows central low-attenuation structures. Excretory phase images (B) show that
the structures do not fill with excreted contrast and represent parapelvic cysts.

often show whether hydronephrosis is present. Other- before image acquisition. The length of the delay
wise, delayed axial images of the kidneys may be mostly depends on the scanner; multidetector-row
obtained and clearly differentiate parapelvic cysts scanners need the slightly longer delay to prevent
from the collecting system (Fig. 4). Delayed images premature imaging of the abdominal organs. In
also may be helpful for patients with an obstructing MDCT, the scans are performed more quickly. The
stone and moderate or large amounts of peripelvic timing used for single-detector CT may yield images
fluid to evaluate for calyceal rupture. that are too early for optimal evaluation.
Active extravasation of arterial or venous contrast
Trauma and general evaluation may be detected during the nephrographic phase as a
collection or linear track of dense contrast emanating
A common indication for CT, which includes renal from the renal parenchyma or renal hilum [8,9].
evaluation, is blunt abdominal trauma. Helical CT is Delayed images may be useful in the setting of
the modality of choice in the evaluation for renal trauma if there is renal laceration or perinephric fluid
laceration or contusion. Indications commonly are to suggest hematoma or urinoma. The delay is
gross hematuria or microscopic hematuria with hypo- generally performed approximately 10 minutes after
tension or additional signs of abdominal hemorrhage. contrast injection. Contiguous 5-mm thick images are
Faster scan acquisition is beneficial to reduce motion usually adequate in this portion of the examination.
artifacts in a noncompliant or obtunded patient. Delayed or excretory images may detect renal hemor-
Breathing misregistration also is reduced in rapid rhage or urinoma that is not visible on routine images
image acquisition. Images with severe motion can (Fig. 5) [10].
be reconstructed using part of the helix to determine
whether a finding is artifact or a true injury. This is
rarely necessary, however, in the urologic system. Renal mass imaging
Helical 5-mm images of the abdomen and pelvis
with a 1.5 pitch are performed at the authors’ insti- The most common nonemergent indication for
tution for blunt abdominal trauma. IV contrast is renal CT at the authors’ institution involves evalua-
routinely administered using 125- to 150-mL low- tion or staging of a renal mass. The mass may be
osmolar contrast at 2 to 4 mL/second rate. IV access symptomatic or one of the increasing number of
is preferred in the antecubital fossa, but hand or lower incidental findings detected as more CTs are being
extremity venous accesses may be used if necessary. performed. Multiphase imaging in a patient with renal
After injection, there is a 60- to 80-second delay mass can serve one of two broad purposes: charac-
M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875 869

Fig. 5. Contrasted CT through the level of the kidneys in a trauma patient. (A) Portal phase shows perinephric fluid, possibly
hemorrhage or urine. (B) Delayed images show dense contrast leaking from the collecting system into the perinephric space
confirming urinoma.

terization of the renal lesion, or staging and detection standard for unenhanced images. Subsequent images
of metastatic disease. after IV contrast are performed in a similar manner
With regard to renal mass characterization, the after 70- to 90-second delay after injection.
typical protocol includes unenhanced and contrasted If staging of a known mass is desired, three-phase
portal and nephrographic phase images (Fig. 6). CT is performed through the liver and kidneys. Occa-
Unenhanced images are performed to detect calcifi- sionally, delayed images of the liver may be performed
cations within the lesion. Also, they provide a base- to help characterize an indeterminate liver lesion. The
line density to allow evaluation of enhancement. At typical arterial phase images begin 25 seconds after
the authors’ institution, 5-mm slice thickness is the start of IV contrast injection. The timing can also be

Fig. 6. Multiphase CT through the level of the kidneys performed for renal mass detection. (A) Precontrast images through the
level of the right kidney show mildly hypodense region in the right kidney. (B) Postcontrast images through the same level show
enhancement of a focal lesion in the region of precontrast hypodensity. Subsequent pathology confirmed renal cell carcinoma.
870 M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875

adjusted using a bolus tracking method or small timing as 1-mm images may be performed to allow coronal
bolus. The portal phase starts 70 to 90 seconds after reconstruction or maximum intensity projection
injection for hepatic imaging. images of the collecting systems and ureters. The
The nephrographic phase of the kidneys occurs optimal delay of imaging after start of contrast infusion
80 to 180 seconds after injection [11]. Some authors is 5 to 10 minutes. A postinjection 250-mL drip
suggest timing of renal images at least 100 seconds to infusion of saline before delays can be used to distend
ensure homogenous enhancement during nephro- the ureters [16]. External compression over the pelvis
graphic phase [12,13]. Others have suggested a may help distend the ureters and collecting systems
120- to 150-second delay [14]. The detection of renal [17,18].
masses is reportedly improved in the nephrographic If a dense renal mass is detected on routine CT,
phase relative to the earlier corticomedullary phase there may be no precontrast images to calculate
[12,15]. A small lesion can be detected that may enhancement. In these cases, delayed images can
blend with the cortex on corticomedullary images show de-enhancement of a renal tumor (Fig. 8).
(Fig. 7). On nephrographic phase images, homo- Vascular renal masses, such as renal cell carcinoma,
genous enhancement of the renal veins is also useful decrease in density on delayed images. At least one
to evaluate for venous invasion. There is only occa- article has shown benefit in delayed images, from as
sionally a benefit to further delayed images of the soon as 30 minutes to 4 hours, to evaluate whether
kidneys or liver in these patients. the lesion has vascularity. The authors suggest that a
Delayed images may also be performed after con- decrease of 15 HU or more is consistent with tumor.
trast is excreted into the collecting systems. The Alternatively, a hyperdense renal cyst shows no
excretory phase begins 3 to 5 minutes after contrast change in density between corticomedullary and
injection. These images may be useful in the evalua- delayed-phase images [19].
tion of central renal masses, whether renal cell carci- CT angiography of the kidneys may be specifically
noma or transitional cell carcinoma (TCC). Filling of requested to evaluate the renal vasculature or the
the collecting systems allows detection of distortion of relationship of vasculature to tumors. This can be
the calyces or renal pelvis. Extension of tumor along extremely important for planning of laparoscopic or
the urothelium can suggest TCC as the etiology of a limited-incision nephrectomy or partial nephrectomy.
mass because renal cell carcinoma does not usually In the authors’ experience, CTA has been very useful
extend along the collecting system or ureter. Three- in preoperative evaluation of tumors within horseshoe
dimensional or multiplane reconstructions can be kidneys. The vessels are often distorted and there is
helpful in these examinations. Axial images as thin often variable vascular supply to the central portions of

Fig. 7. Contrasted CT during corticomedullary phase (A) shows no focal abnormality. Repeat CT through the same level (B) with
nephrographic timing demonstrates small enhancing mass, which was confirmed as renal cell carcinoma.
M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875 871

Fig. 8. De-enhancement of incidentally discovered renal mass in a patient in whom precontrast images were not obtained.
(A) Contrasted CT at the level of the kidney shows hyperdense lesion measuring 117 HU. (B) Subsequent short-interval delayed
image at the same level measures 91 HU in the same region, confirming vascularity of the lesion.

the fused kidney. Helical 1.2- to 2.5-mm slices are At the authors’ institution they perform a postscan
obtained using arterial phase timing. Timing is per- scout image to evaluate for ureteral duplication. Other
formed using a bolus tracking system or timing bolus authors have suggested that the resolution of the CT
to ensure optimal opacification of the arteries. This is scout is insufficient to exclude medullary sponge
especially important in patients with abnormal cardiac kidney or papillary necrosis, and they recommend
output [20] or young hyperdynamic patients. postcontrast conventional radiography [21].

Renal donor evaluation Crossing vessels evaluation

Up to five series have been used in the evaluation An increasing use of minimally invasive proce-
of patients for renal donation [21]. In the evaluation dures on the urologic system has brought potential
of potential renal donors at the authors’ institution, new complications, whether because of limited visua-
however, three series are typically obtained to reduce lization caused by smaller incisions in open proce-
radiation dosage to the healthy donor. Noncontrast dures or endoscopic limitations. Endoscopic repair of
images are initially obtained to evaluate for nephro- ureteropelvic junction obstruction can be performed
lithiasis and as baseline for measurement of enhance- using a blind incision through the ureter wall. A
ment in case a renal lesion is detected. The images potential complication occurs if the incision encoun-
also permit characterization of any adrenal nodules, ters an abnormally positioned renal artery. Abnormal
if present. crossing vessels are documented in up to 50% of the
Intravenous contrast is given at a rate of 3 to population [22] and in greater than 50% of patients
5 mL/second (preferably 5 mL/second), and arterial with ureteropelvic junction obstruction [23]. The
phase images are obtained to evaluate renal vascula- vessels are usually anterior to the ureter. Ureterotomy
ture and allow three-dimensional reconstructions. The is typically performed in the posterior aspect of the
authors use a MDCT with 1.25-mm images obtained ureter wall to minimize the risk [24]. Posterior
after a delay determined by bolus tracking. Late portal crossing vessels are common, however, representing
or nephrographic-phase images allow detection of 6 of 13 crossing vessels in cases of ureteropelvic
renal masses, parenchymal abnormalities, cysts, junction obstruction in one series [23], and may result
and extrarenal abnormalities. For these images, helical in postprocedure hemorrhage. CTA is very accurate
5-mm images are obtained after a 90- to 180-second in detecting crossing renal arteries and can help
delay from the time of injection. prevent such problems.
872 M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875

Radiation dosage issues Several other situations, however, should trigger


additional effort to reduce dose. In young patients,
Single detector versus MDCT radiation dosage especially young women, who are expected to have
repeated CT studies in the future, extra effort should
Although many parameters and terms have be taken to reduce the dose to the minimum possible.
changed since the advent of multislice CT, the cal- These patients may include complicated pancreatitis
culation of dose is the same for single-slice helical patients, chronic renal stone formers, or young
CT as it is for multislice CT. Initial estimates showed patients with treated tumors who have good progno-
that MDCT results in higher radiation dosages if sis but need repeated follow-up to monitor for recur-
techniques similar to SDCT are chosen. Lower milli- rence of tumor.
ampere and kilovolt (peak) settings, however, yield Several articles have discussed low-dose tech-
similar quality images with similar dosimetry. Because niques for detection of renal calculi with good clinical
there are multiple rows of detectors, the penumbra of results in adults. Hamm et al [27] showed sensitivity
radiation for one slice is beneficial to the adjacent slice and specificity of 96% and 97%, respectively, using
in the central detectors. Although the radiation dose 120 kV, 70 mA, 5-mm collimation, pitch 2, with
experienced by the patient from the radiation beam is 5-mm reconstructions. This technique reduced radia-
similar to single-detector CT, the MDCT makes more tion exposure by 50% to 1.50 mSv, which was com-
efficient use of the total radiation produced by the tube parable with exposure for excretory urography [27].
because it gets multiple slices for the same tube current This was similar to results noted by Liu et al [28] using
and time. a protocol with 7-mm slices and a slightly higher
technique yielding an exposure of 2.8 mSv. The
Low-dose renal calculus authors noted that the technique might be limited in
obese patients [27]. Tack et al [29] have recently sug-
Radiation exposure has moved to the forefront in gested 30-mA technique with further focused imaging
the national media and radiologic community. As can be accurate, but as noted in an accompanying
diagnostic imaging, mainly CT, has become more commentary, a 60-mA technique may eventually be
commonplace, it has become one of the largest man- determined as more appropriate for adequate diagnosis
made contributors to population radiation exposure. of urinary or unexpected nonurinary etiologies in adult
At the authors’ institution, over 12 million CT images patients in the United States [30].
have been generated since January 1999. As the Because dosimetry is similar for single-detector or
amount of CT imaging increases, there are concerns multidetector CT, the methods to minimize the pa-
of deleterious effects that this radiation could have on tient radiation dose are similar in both systems. The
society and individuals regardless of outcome of deposited radiation correlates directly in a propor-
persistent debate whether threshold effects exist with tional manner to the milliampere used. Double the
respect to CT levels of radiation dosage. It is essential milliampere results in double the patient dose, when
to avoid unnecessary exposure and reduce the radia- all else remains equal. Even small reductions in kilo-
tion exposure to the lowest possible levels while still volt (peak) significantly reduce image quality without
producing diagnostic image quality. significantly reducing patient radiation. Dose reduc-
In pediatric populations, CT is a significant source tion is usually best achieved through reduction of the
of radiation. Pediatric CT protocols for detection of milliampere rather than kilovolt (peak). Typically, the
urinary tract calculi vary by institution. The best ways milliampere is approximately 200 to 240 for 5-mm
to reduce dose include a reduction in milliampere and thick images in a standard CT of an adult patient. In
increase in pitch [25]. In one study, three techniques pregnant patients, the authors often reduce the milli-
for renal calculi chose 3- to 5-mm collimation, 180 to ampere to 60 to 100, depending on patient body
280 mA, pitch 1:1 to 2:1, and 120 kVp. These values habitus. Increasing the pitch can significantly reduce
yielded estimated ovarian dose of 0.31 to 1.07 rad to dose to the patient. The effective slice thickness in
the child. It is noted that by reducing the milliampere the z-axis may be increased, however, and this
to 100 and using 2:1 pitch the estimated ovarian dose could reduce sensitivity for small lesions, such as
is 0.15 to 0.26 rad, a significantly reduced dose that ureteral calculi.
provided diagnostic image quality [26]. Commonly, a
pitch of 1.5 or 2 may be used in pediatric patients Pregnant patient evaluation
without loss of diagnostic information [25].
Dose reduction is most commonly discussed One of the most effective methods of dose reduc-
in pregnant and pediatric patients, as is justified. tion is the appropriate selection of alternate modali-
M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875 873

ties that do not use ionizing radiation. The kidneys because of the significantly higher sensitivity and
often are well demonstrated by ultrasound, and ultra- specificity of CT.
sound should be considered as a primary examination
in the evaluation of renal abnormalities in pregnant Pediatric CT issues
patients. The latest ultrasound machines have many
techniques to aid in organ visualization, such as Dose reduction
harmonic and pulse inversion imaging. Ultrasound Pediatric renal CT is most commonly performed
usually does not image the ureters well and may miss to evaluate for renal mass, such as Wilms’ tumor. CT
smaller stones or tumors in the kidneys. is an excellent modality to evaluate for adenopathy or
MR imaging is an excellent alternative modality renal vein involvement. Distant metastases are also
for renal evaluation despite its expense and sensitivity detectable by CT. For infants, the radiographic tech-
to motion artifact. In patients who are able to have the nique is significantly lower than for adults because of
study, the kidneys can be quite well evaluated. the lower body mass of the patient. Standard milli-
Additionally, there is less risk of nephrotoxicity, ampere and kilovolt (peak) values for children vary
which is a concern when performing CT of patients by the size of the child. For an infant, 60 to 70 mA
with renal insufficiency. MR angiography provides and 120 kVp have been recommended. For a larger
excellent evaluation of the abdominal and pelvic child who weighs 60 to 79 lb, 100 mA and 120 kVp
arterial systems. are adequate. Adult protocols using 220 mA, how-
In pregnancy, as in all patients, the principle of ever, are not appropriate for pediatric patients [25].
‘‘as low as reasonably achievable’’ is followed. Still, The timing of imaging for standard abdominal eval-
there are certain times in the gestation of a pregnancy uation is earlier than in adults (50 versus 70 seconds)
that are more susceptible to the effects of radiation. because of the faster circulation time. The volume of
The period of organ morphologic development in the IV contrast is 1 mL/lb or 2 mL/kg given at a rate of
first trimester is of special concern. The third trimes- 1 to 3 mL/second or by hand injection.
ter is the period of least sensitivity to the effects of
radiation, but dose reduction is still crucial in these Alternative modalities to limit radiation exposure
patients. In the acute setting, such as after severe In children, there are several alternative imaging
trauma, where the life of the mother is at significant methods for evaluation of renal disease. Because of
risk, the radiation dose reduction should be secondary the small size of the patients, excellent sonographic
to diagnostic image quality. In these circumstances penetration is possible. The kidneys are smaller and
CT may be performed without hesitation because the closer to the skin than in adults, so higher-frequency
death of the mother from a missed injury would transducers may penetrate to the kidneys well and can
likewise result in death of the pregnancy. provide excellent spatial resolution and image quality.
After trauma, the CT most often requested in MR imaging is an alternative, but it may require
pregnant patients is for the evaluation of renal colic. sedation in younger pediatric patients.
Often there are hemodynamic changes of pregnancy,
and many women may have episodes of dehydration,
predisposing to renal calculi. The evaluation of renal New concepts
colic in a pregnant patient is a difficult diagnostic
situation. There may be hydronephrosis of pregnancy Volumetric acquisition: plate detectors
complicating the imaging picture. Also, the enlarged
uterus often displaces the ureters. In these patients, Although makers of CT scanners continue to
low-dose noncontrast CT is usually performed if a make incremental improvements in number of rows
calculus is not evident on conventional abdominal of detectors, the next major transition will likely
radiograph. The dose should be reduced as much as include image plate detector technology in CT. There
possible, but care is taken not to perform a non- have been early articles using video fluoroscopy or
diagnostic study, which is a needless radiation expo- image intensifiers published on the developing tech-
sure. A single test slice may be performed through the nology [31,32] and it may greatly improve the spatial
upper abdomen above the uterus to evaluate whether resolution of CT for imaging of very small structures
a reduced technique yields too much noise to detect a within the body. Characterization of smaller feeding
small stone. Low-dose noncontrast CT has all but vessels will be possible. Less volume averaging or
replaced limited IV pyelogram in stone evaluation at pixelation of the images should improve image qual-
the authors’ institution even in pregnant patients ity. Because there will be no spacing between detec-
874 M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875

tors, less radiation should remain uncollected in the and vascular anatomy is well suited to CT techniques.
image acquisition. Subtle adjustments in the technical parameters and
timing of the study, however, can optimize the
Molecular imaging applications evaluation based on the clinical setting. As CT is
more widely used, often repeatedly on an individual
There are no reimbursed routine indications for patient, radiation exposure must be minimized while
molecular imaging for the initial detection of tumors still obtaining diagnostic image quality.
in the kidneys. Although many tumors, such as
lymphoma, can be detected and characterized easily
with fluorine-18-fluorodeoxyglucose positron emis-
sion tomography, renal cell carcinoma has not shown Acknowledgments
as much promise with this agent. Positron emission
tomography has high positive predictive value in this The authors thank Trish Dobbs for her assistance
setting, however, and may be used for characterization with manuscript preparation, and Anthony Zagar for
of lesions detected by other modalities [33]. This photographic assistance.
technique may be used for evaluation of residual
tumor after nephrectomy. It also may be used to
evaluate response to therapy for metastatic disease
References
[33]. Similar benefits may be seen for tumors of the
urinary bladder, but the applications in prostate cancer [1] Polacin A, Kalender WA, Marchal G. Evaluation of
evaluation may be limited [33]. It is hoped that section sensitivity profiles and image noise in spiral
additional agents will move into general practice to CT. Radiology 1992;185:29 – 35.
enable physicians to characterize the metastatic poten- [2] Megibow AJ, Jacob G, Heiken JP, Paulson EK, Hopper
tial of renal lesions. Positron emission tomography CT KD, Sica G, et al. Quantitative and qualitative evalua-
imaging is growing in usage and combines high spatial tion of volume of low osmolarity contrast medium
resolution images of the anatomy by CT with the needed for routine helical abdominal CT. AJR Am J
functional images of positron emission tomography. Roentgenol 2001;176:583 – 9.
The CT technique used for attenuation correction at [3] Williamson EE, McKinney JM. Assessing the ade-
quacy of peripherally inserted central catheters for
the authors’ institution generally consists of 130 kVp
power injection of intravenous contrast agents for
and 90 mA with 0.8-second helix in this situation. CT. J Comput Assist Tomogr 2001;25:932 – 7.
[4] Carlson JE, Hedlund LJ, Trenkner SW, Ritenour R,
Compression for distention Halvorsen Jr RA. Safety considerations in the power
injection of contrast media via central venous catheters
Another variant of standard CT uses a technique during computed tomographic examinations. Invest
commonly used in IV urography. In IV urograms, the Radiol 1992;27:337 – 40.
initial images of the collecting systems are obtained [5] Herts BR, O’Malley CM, Wirth SL, Lieber ML, Pohl-
with improved distention of the proximal collecting man B. Power injection of contrast media using central
systems by using compression pads placed over the venous catheters: feasibility, safety, and efficacy. AJR
Am J Roentgenol 2001;176:447 – 53.
pelvic brim. The same concept can be applied to CT
[6] Rubin GD. Techniques for performing multidetector-
for the imaging of the renal collecting systems in the row computed tomographic angiography. Tech Vasc
evaluation for possible TCC. The technique may also Interv Radiol 2001;4:2 – 14.
be helpful in determining whether a renal cell carci- [7] Gottlieb RH, La TC, Erturk EN, Sotack JL, Voci SL,
noma extends to the urothelium for surgical planning. Holloway RG, et al. CT in detecting urinary tract cal-
Compression over the ureters allows increased dis- culi: influence on patient imaging and clinical out-
tention of the proximal collecting system. Release of comes. Radiology 2002;225:441 – 9.
compression may improve filling of the ureters [8] Shanmuganathan K, Mirvis SE, Sover ER. Value of
[18,34] for evaluation of an urothelial mass or clot. contrast-enhanced CT in detecting active hemorrhage
in patients with blunt abdominal or pelvic trauma. AJR
Am J Roentgenol 1993;161:65 – 9.
[9] Jeffrey RB, Cardoza JD, Olcott EW. Detection of
Summary active intraabdominal arterial hemorrhage: value of
dynamic contrast-enhanced CT. AJR Am J Roentgenol
CT is a robust, rapid means of evaluation for a 1991;156:725 – 9.
wide spectrum of urologic disorders. The evaluation [10] Yuh BI, Cohan RH. Different phases of renal enhance-
of renal trauma, urologic malignancy, urolithiasis, ment: role in detecting and characterizing renal masses
M.E. Lockhart, J.K. Smith / Radiol Clin N Am 41 (2003) 863–875 875

during helical CT. AJR Am J Roentgenol 1999;173: [22] Quillin SP, Brink JA, Heiken JP, Siegel CL, McClennan
747 – 55. BL, Clayman RV. Helical (spiral) CT angiography for
[11] Sheth S, Scatarige JC, Horton KM, Corl FM, Fishman identification of crossing vessels at the ureteropelvic
EK. Current concepts in the diagnosis and manage- junction. AJR Am J Roentgenol 1996;166:1125 – 30.
ment of renal cell carcinoma: role of multidetector [23] Farrés MT, Pedron P, Gattegno B, Haab F, Tigui M,
CT and three-dimensional CT. Radiographics 2001;21: Carette MF, et al. Helical CT and 3D reconstruction of
S237 – 54. ureteropelvic junction obstruction: accuracy in detec-
[12] Cohan RH, Sherman LS, Korobkin M, Bass JC, Fran- tion of crossing vessels. J Comput Assist Tomogr 1998;
cis IR. Renal masses: assessment of corticomedullary- 22:300 – 3.
phase and nephrographic-phase CT scans. Radiology [24] Streem SB, Geisinger MA. Prevention and manage-
1995;196:445 – 51. ment of hemorrhage associated with cautery wire bal-
[13] Silverman SG, Lee BY, Seltzer SE, Bloom DA, Corless loon incision of ureteropelvic junction obstruction.
DF, Adams DF. Small (= 3 cm) renal masses: correlation J Urol 1995;153:1904 – 6.
of spiral CT features and pathologic findings. AJR Am J [25] Donnelly LF, Frush DP. Cross-sectional imaging of
Roentgenol 1994;163:597 – 605. abnormalities of the abdominal wall in pediatric pa-
[14] Birnbaum BA, Jacobs JE, Ramchandani P. Multiphasic tients. AJR Am J Roentgenol 2001;176:1233 – 9.
renal CT: comparison of renal mass enhancement during [26] Strouse PJ, Bates DG, Bloom DA, Goodsitt MM. Non-
the corticomedullary and nephrographic phases. Radiol- contrast thin-section helical CT of urinary tract calculi
ogy 1996;200:753 – 8. in children. Pediatr Radiol 2002;32:326 – 32.
[15] Szolar DH, Kammerhuber F, Altziebler S, Tillich M, [27] Hamm M, Knopfle E, Wartenberg S, Wawroschek F,
Breinl E, Fotter R, et al. Multiphasic helical CT of the Weckermann D, Harzmann R. Low dose unenhanced
kidney: increased conspicuity for detection and charac- helical computerized tomography for the evaluation of
terization of small (< 3-cm) renal masses. Radiology acute flank pain. J Urol 2002;167:1687 – 91.
1997;202:211 – 7. [28] Liu W, Esler SJ, Kenny BJ, Goh RH, Rainbow AJ,
[16] McTavish JD, Jinzaki M, Zou KH, Nawfel RD, Silver- Stevenson GW. Low-dose nonenhanced helical CT of
man SG. Multi-detector row CT urography: compari- renal colic: assessment of ureteric stone detection and
son of strategies for depicting the normal urinary measurement of effective dose equivalent. Radiology
collecting system. Radiology 2002;225:783 – 90. 2000;215:51 – 4.
[17] McNicholas MMJ, Raptopoulos VD, Schwartz R, [29] Tack D, Sourtzis S, Delpierre I, de Maertelaer V, Ge-
Sheiman RG, Zormpala A, Prassopoulos PK, et al. venois PA. Low-dose unenhanced multidetector CT of
Excretory phase CT urography for opacification of patients with suspected renal colic. AJR Am J Roent-
the urinary collecting system. AJR Am J Roentgenol genol 2003;180:305 – 11.
1998;170:1261 – 7. [30] Katz DS, Venkataramanan N, Napel S, Sommer FG.
[18] Caoili EM, Cohan RH, Korobkin M, Platt JF, Francis Can low-dose unenhanced multidetector CT be used
IR, Faerber GJ, et al. Urinary tract abnormalities: ini- for routine evaluation of suspected renal colic. AJR
tial experience with multi-detector row CT urography. Am J Roentgenol 2003;180:313 – 5.
Radiology 2002;222:353 – 60. [31] Ning R, Kruger RA. Image intensifier-based computer
[19] Macari M, Bosniak MA. Delayed CT to evaluate renal tomography volume scanner for angiography. Acad
masses incidentally discovered at contrast-enhanced Radiol 1996;3:344 – 50.
CT: demonstration of vascularity with deenhancement. [32] Endo M, Yoshida K, Kamagata N, Satoh K, Okazaki T,
Radiology 1999;213:674 – 80. Hattori Y, et al. Development of a 3D CT-scanner using
[20] Birnbaum BA, Jacobs JE, Langlotz CP, Rmchandani P. a cone beam and video-fluoroscopic system. Radiat
Assessment of a bolus-tracking technique in helical Med 1998;16:7 – 12.
renal CT to optimize nephrographic phase imaging. [33] Shvarts O, Han KR, Seltzer M, Pantuck AJ, Bellde-
Radiology 1999;211:87 – 94. grun AS. Positron emission tomography in urologic
[21] Rydberg J, Kopecky KK, Tann M, Persohn SA, Leap- oncology. Cancer Control 2002;9:335 – 42.
man SB, Filo RS, et al. Evaluation of prospective [34] Caoili EM, Bude RO, Higgins EJ, Hoff DL, Nghiem
living renal donors for laparoscopic nephrectomy with HV. Evaluation of sonographically guided percuta-
multisection CT: the marriage of minimally invasive neous core biopsy of renal masses. AJR Am J Roent-
imaging with minimally invasive surgery. Radio- genol 2002;179:373 – 8.
graphics 2001;21:S223 – 36.
Radiol Clin N Am 41 (2003) 877 – 907

MR techniques for renal imaging


Jingbo Zhang, MD, Ivan Pedrosa, MD, Neil M. Rofsky, MD*
Department of Radiology, Beth Israel Deaconess Medical Center, Shapiro 4 Clinical Center, 330 Brookline Avenue, Boston,
MA 02215, USA

Cross-sectional imaging plays a critical role in Overview: role of MR imaging in genitourologic


detection and work-up of renal pathologies. MR evaluations
imaging provides exquisite, versatile, and unique soft
tissue contrast, and allows for an effective evaluation Renal mass evaluation
of a wide range of renal disorders. MR imaging
techniques with rapid acquisition times can bypass The detection of kidney tumors has substantially
many of the motion artifacts that previously posed progressed over the last 15 years because of improved
limitations to abdominal MR imaging and are now imaging techniques [4], which have led to earlier
widely available. MR imaging is especially attractive detection and improved survival. The incidence of
in assessing renal-related disorders in children, in asymptomatic renal masses has increased up to 30%
women of childbearing age, and in patients with during this time [5]. MR imaging has long played an
renal insufficiency or renal allografts. This appeal adjunctive role for the characterization of renal
is the result of the lack of exposure to ionizing masses that were indeterminate by ultrasound and
radiation and the safety profile of the Food and CT [3,6,7].
Drug Administration – approved gadolinium contrast Recent improvements in MR imaging have
agents. The latter includes an extraordinarily low rate changed the playing field. Taken together with some
of anaphylactoid reactions and, in particular, the pitfalls that have been recognized with CT, such as
ability to be used safely in the setting of pre-existent pseudoenhancement [8,9], MR imaging has emerged
renal insufficiency [1 – 3]. Renal MR imaging has as a nearly ideal technique for the detection, diagno-
now evolved as an alternative or complementary sis, staging, and preoperative evaluation of renal
imaging modality to ultrasound, excretory urography, masses. Crescents of normal renal cortex separating
and CT. adjacent multiple cysts that appear as single lesions
This article reviews the currently available with thick, enhancing septae on CT can be clarified
MR imaging strategies for the evaluation of the with the multiplanar capability, enhancement fea-
renal-related disorders, and provides specific recom- tures, and T2 signal characteristics of MR imaging
mendations to generate images with consistent diag- [7]. MR imaging also can stage the tumor at the time
nostic efficacy. of diagnosis, which facilitates an assessment of
prognosis and surgical planning. Parenchymal
lesions, venous extension, and adenopathy are all
clearly depicted.
The increased number of incidentally detected
renal masses, particularly smaller masses, has had
* Corresponding author. a substantial impact on the treatment of renal cell
E-mail address: nrofsky@caregroup.harvard.edu carcinoma (RCC) over the last decade. The increased
(N.M. Rofsky). reliance on nephron-sparing surgery, including less-

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00069-1
878 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

invasive laparoscopic procedures, has maximized the images of the renal arteries can be obtained, with
amount of remnant renal tissue without sacrificing atherosclerotic lesions, occlusions, and aneurysms
cancer control [10]. Indeed, studies suggest that well depicted.
partial nephrectomy or wedge resection of local- Images similar in appearance to digital subtraction
ized RCC yields cure rates similar to those obtained angiography are generated with the use of postpro-
with radical surgery [11], providing a long-term func- cessing techniques, such as maximum intensity pro-
tional advantage. jection (MIP) and volume rendering (VR). The spatial
In this context, the imaging evaluation is particu- resolution of MR angiography is lower than that of
larly important when planning complex nephron- digital subtraction angiography, but this is balanced
sparing treatments. A comprehensive gadolinium- by its documented efficacy and safety. Indeed, the
enhanced multiplanar MR imaging study integrates consistently excellent correlation of MR angiography
all of the necessary information previously obtained with conventional angiography has generated an
by conventional CT, angiography, venography, and increased reliance on this technique and may even
pyelography into a single preoperative test. Because replace arteriography in most patients with suspected
the growth rate of small renal tumors is slow or renal artery stenosis [18,21 – 35]. Furthermore, MR
nonexistent [12,13], an option for a watchful waiting angiography can be the only suitable option for
approach to small renal tumors has been suggested certain patients referred to assess a vascular etiology
[13,14]. MR imaging can serve as an effective tool for renal insufficiency.
for follow-up of these small masses. The cross-sectional volumetric nature of con-
trast-enhanced MR angiography affords advantages
Urothelial tumor over conventional catheter angiography [19,36,37].
Although conventional angiography has been con-
The urothelium is a target tissue for carcinogens sidered the gold standard, its limited projectional
that can lead to the development of transitional cell views may cause obscuration of the proximal renal
carcinomas (TCCs). The urinary bladder is the most arteries [38] and underestimation of en face ather-
common site of TCC. Any urothelial surface is sus- omatous plaques. The volumetric MR angiography
ceptible, however, including the intrarenal collecting enables true three-dimensional imaging, demon-
system and the renal pelvis. strating perspectives of renal artery stenoses that are
On MR imaging studies, urothelial tumors present unattainable with the limited number of two-dimen-
as enhancing, irregular fixed masses arising from any sional projectional views inherent to conventional
urothelial surface, or focal wall thickening, either angiography (Fig. 1) [19,21]. An additional benefit
eccentric or circumferential. TCC is usually confined of MR angiography is in detecting incidental but sig-
to the collecting system lumina, but lesions can nificant pathologies, including parenchymal lesions
extend into the renal parenchyma, typically in an in the intra-abdominal organs and other potential
infiltrative pattern that preserves the reniform shape. causes for renal insufficiency or hypertension (eg,
The relatively slow growth of ureteral TCC allows for adrenal masses) [39 – 41].
gradual expansion of the ureteral lumen and is less A unique capability of MR angiography is its
likely to produce acute symptoms [15]. MR urogra- ability to supplement a vascular display with hemo-
phy has been performed to demonstrate the dilated dynamic and functional adjuncts [21,42 – 45]. This
urinary collecting systems associated with urothelial capability is particularly important for determining
tumors [16]. Technical details regarding MR urogra- the likelihood of achieving a favorable response to
phy can be found elsewhere in this issue. revascularization [46]. The simultaneous acquisition
of angiographic images with time-resolved reno-
Renal artery evaluation graphic data can provide a quantitative measurement
of renal perfusion [21,47 – 50]. Associated findings,
During the past decade, MR angiography has such as poststenotic dilatation, delayed renal en-
evolved from an experimental technique into the hancement, and reduced renal parenchymal mass,
modality of choice for the noninvasive evaluation help to determine the hemodynamic significance of
of renovascular disease [17] and a serious alternative a renal artery stenosis [51,52].
to conventional angiography. The recent widespread Quantitative assessments of renal function have
application of MR angiography for these indications been pursued including MR phase-contrast flow
has been driven primarily by the advent of three- measurements, quantitative perfusion measurements
dimensional contrast-enhanced MR angiography, a with intravascular contrast agents, MR renography,
robust technique with high accuracy [18 – 20]. Superb and excreting contrast evaluations. By facilitating an
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 879

Fig. 1. Renal artery stenosis in 63-year-old man with hypertension. (A) Coronal maximum intensity projections (MIP)
reconstruction of a three-dimensional fat-saturated T1-weighted gradient echo acquisition (TR = 3.8, TE = 1.9, FA = 25, slice
thickness = 4 mm, before interpolation) during the arterial phase after administration of a single dose of gadolinium (0.1 mmol/kg
body weight). There is mild stenosis in the proximal right renal artery (arrow). Note the atheromatous changes in the infrarenal
abdominal aorta (arrowheads). (B) Oblique axial view of the same MIP reconstruction as Fig. 1A shows a significant stenosis in
the proximal right renal artery secondary to an atheromatous plaque in the anterior wall of this vessel (arrow). Volumetric
acquisitions allow for MIP reconstructions that can be displayed in virtually any spatial orientation to demonstrate better the area
of interest. (C) Coronal image from a conventional angiogram in the same patient demonstrates only moderate stenosis of the
proximal right renal artery (arrow). Limited available projections in conventional angiography make difficult visualization of en
face plaques in the renal artery. Conventional angiography confirms the atheromatous changes in the infrarenal aorta with
ulcerated plaques (arrowheads).

assessment of renal blood flow, perfusion, glomeru- Current applications of renal MR angiography
lar filtration rate, and functional impact in response range from detection of renal artery stenosis, planning
to pharmacologic challenge renal excretion, these of renal revascularization, to preoperative evaluation
techniques can improve diagnostic specificity [42, of potential transplant donors and recipients [57].
43,48,53 – 56]. Contrast-enhanced MR angiography also is a reli-
880 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

able method in identifying postoperative vascular determining the superior extent of tumor thrombus in
complications and perfusion defects after renal re- the inferior vena cava, especially in the region of the
vascularization and in kidney allografts following right atrium [64,65].
transplantation [19,57 – 62]. Although reliable fol- This information potentially impacts the surgical
low-up can be obtained in patients following angio- approach in cases where CT or ultrasound is equivo-
plasty, the results following stent placement can be cal [66 – 68]. The same technique used for three-
plagued by artificial signal loss from certain stent dimensional gadolinium-enhanced MR angiography
materials [63]. is ideal for noninvasive evaluation of the renal veins
and inferior vena cava (Fig. 2). Multiplanar refor-
Renal vein evaluation mations from three-dimensional data sets are often
helpful in delineating tumor extent. Contrast-en-
The assessment of the renal veins is an important hanced study with subtraction helps to differentiate
component of the work-up of a patient with RCC. bland thrombus from enhancing tumor thrombus
The demonstration of tumor extension into the in- (Fig. 3).
ferior vena cava or into the right atrium affects
treatment planning and the surgical specialties that
potentially assist the urologist. MR imaging has Imaging options
demonstrated an excellent depiction of tumor throm-
bus. It has been shown as a favorable technique for First discussed are broad imaging options. Then,
further details are provided guiding the selection and
recommendation of the possible approaches for opti-
mizing renal imaging. Tables 1 and 2 include the
particulars to help the user select specific MR imag-
ing parameters for robust diagnostic efficacy.

Breathhold imaging

In cooperative patients the most vexing problems


associated with MR imaging arise from artifacts
secondary to physiologic motion: respirations, car-
diac pulsations, and bowel peristalsis. Fast imaging
and single-shot pulse sequences in conjunction with
breathholding are the most effective techniques to
eliminate respiratory artifacts.
Suspended respiration is most reproducible in
end-expiration, a key consideration when subtraction
postprocessing is needed. End-expiratory breathhold-
ing is maximized by a brief coaching session in
which the patient is informed of the importance of
avoiding extremes in respiratory efforts and the goal
Fig. 2. Left renal cell carcinoma with vein thrombosis. In the of achieving a constant lung volume at the end of
presence of left renal vein thrombosis MR images provide each expiration. The use of hyperventilatory prepa-
critical information that affect the decision for the surgical rations can facilitate the breathhold procedure. The
approach. The superior mesenteric artery (SMA) is used as authors have found that two cycles of the command
an anatomic landmark for the midline. If the thrombus is ‘‘breath in, breath out’’ yield good results.
proximal to the SMA, a left flank approach is used. If An additional benefit of the coaching session is
the thrombus extends beyond the SMA, a midline inci- the ability to identify patients with the most limited
sion is preferred. Coronal three-dimensional fat-saturated capacity to sustain a breathhold during a practice set
T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA =
of commands. In these patients, the use of a nasal
12, slice thickness = 4 mm, before interpolation) during the
portal venous phase after administration of gadolinium
cannula to administer oxygen greatly increases the
demonstrates an intraluminal filling defect in the left renal individual’s breathhold ability [69]. Alternatively,
vein (arrow) that does not extend beyond the SMA end-inspiration with or without oxygen supplemen-
(arrowhead). Findings were confirmed at surgery with left tation can be used, and nonbreathhold strategies may
flank approach. be needed.
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 881

Fig. 3. Left papillary renal cell carcinoma with renal vein thrombosis in a 51-year-old man presenting with pulmonary embolism
and stroke. Echocardiogram revealed a patent foramen ovale. (A) Coronal subtracted three-dimensional fat-saturated T1-
weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) demonstrates a diffuse
infiltrative mass in the left kidney (arrows). (B) Coronal half-Fourier single-shot turbo spin echo (HASTE) image (TR = 1100,
TE = 64, FA = 130, slice thickness = 4 mm) of the abdomen shows complete filling of the left renal vein by tumoral thrombus
extending to the level of the inferior vena cava (IVC) (arrow). Note an area of susceptibility artifact immediately adjacent to the
tumor thrombus related to a previously placed IVC filter (white arrow). The SMA also is visualized (arrowhead). Tumor
thrombus was confirmed at surgery partially adhered to the IVC filter. Patient presented 6 months later with edema of both lower
extremities. A repeated MR imaging examination was obtained. (C) Coronal subtracted three-dimensional fat-saturated T1-
weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) confirms the
thrombosis of the IVC. Note the heterogeneous enhancement consistent with tumoral thrombus (arrows) and nonenhancing areas
related to bland thrombus (arrowheads). IVC filter (thick white arrow).
882 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

Table 1
Sequences for renal mass protocol
Sequence Type Goals Additonal notes
1. Scout Multiplanar two-dimensional GRE Localization
2. Dual echo GRE Axial, two-dimensional GRE Characterize adrenal Can help detect susceptibility
lesions; help characterize when blooming noted in later echo
renal lesions
3. Sagittal T1 Three-dimensional GRE with Lesion detection and Can be oblique orientation;
fat suppression characterization separate acquisition of each kidney
4. Coronal T2 Two-dimensional half-Fourier, Lesion detection and Plan off sagittals
single shot through kidneys limited characterization
5. Coronal T1 Three-dimensional GRE with Lesion detection and Precontrast and two postcontrast
fat suppression characterization; Vascular scans (arterial and nephrographic
evaluation phases)
6. Delayed Three-dimensional GRE with Lesion detection and Subtract precontrast from
post-gadolinium fat suppression characterization postcontrast
(repeat sequences 2
and 3)

One option that can be effective but is relatively yields motion-free images. As retroperitoneal struc-
time consuming is the retrospective averaging of tures the kidneys are somewhat restricted in their
individual breathholds [70]. Imaging strategies that motion, and renal images are less apt to be corrupted
are relatively signal poor can be averaged to boost the by subtle movements.
signal-to-noise ratio (SNR) and improve the image Single-shot imaging provides a motion-insensitive
quality [70]. The success of this retrospective averag- strategy because each slice is acquired in less than
ing is dependent on image co-registration and hence, 1 second. The use of suspended respiration is not
reproducible breathhold capability. Because of the required for image quality. When feasible, however, a
time constraints of this technique and the advent of breathhold is recommended because it eliminates the
single-shot sequences, it is less commonly used. misregistration of slices and allows for the anatomy
to be demonstrated in a sequential manner.
Nonbreathhold imaging Magnetization-prepared gradient-echo (MagPrep-
GRE) imaging is quite useful for generating fast
Fast acquisitions motion insensitive T1-weighted images. It allows
When breathhold imaging is not possible, fast for very fast acquisition times on a per-slice basis,
sequences, single-shot imaging, or respiratory cor- eliminating the strict requirement for a breathhold to
rection techniques can be performed. For ventilated achieve motion-free images. In this approach, images
patients, temporary suspension of the respirators often are acquired sequentially, each image requiring less

Table 2
Sequences for renal MRA protocol
Sequence Type Goals Additonal notes
1. Scout Multiplanar 2D GRE Localization
2. Dual echo GRE Axial, two-dimensional GRE Characterize adrenal lesions; Can help detect susceptibility when
help characterize renal lesions blooming noted in later echo
3. Coronal T2 Two-dimensional half-Fourier, Lesion detection and limited
single shot through kidneys characterization
4. High resolution Three-dimensional GRE with Renal vascular evaluation (lesion Precontrast and two postcontrast
axial T1 fat suppression detection and characterization) scans (arterial and nephrographic
phases)
5. High resolution Three-dimensional GRE with Renal vascular evaluation, Precontrast and two postcontrast
coronal T1 fat suppression including iliacs, celiac axis and scans (arterial and nephrographic
SMA origin (lesion detection phases)
and characterization)
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 883

than 1.5 seconds per slice. Pulsation and motion ratory ordered-phase encoding [73] or respiratory
artifacts are largely eliminated [71,72]. gating. Respiratory gating limits data acquisition to
With this approach is the potential for misregis- end-expiration and is successful in restoring sharpness
tration among data sets. This can be seen when and reducing ghost artifacts. Data are collected, how-
attempting to compare in- and out-of-phase Mag- ever, only for a fraction of the respiratory cycle;
PrepGRE images, typically obtained as separate therefore, substantial increases in imaging time are
image sets. This presents a disadvantage compared incurred. Respiratory triggering, however, initiates the
with breathhold dual-echo in- and out-of-phase acquisition of an MR imaging section at a fixed point
images. Furthermore, misregistration is often pres- of the respiratory cycle, restoring sharpness and re-
ent between unenhanced and enhanced sequences, ducing ghosts. Unlike gating, triggering can be used to
making subtraction postprocessing difficult. Another produce an image at any phase of the respiratory cycle,
disadvantage of MagPrepGRE is reduced SNR com- but it requires the use of long repetition times (TR) [74].
pared with traditional T1-weighted breathhold GRE A practical technique that can benefit all the
sequences. Despite the recognized limitations, Mag- previously mentioned approaches is the use of an
PrepGRE imaging is a vital strategy for obtaining abdominal binder, an elastic garment that can be
diagnostic-quality T1-weighted images in those in- wrapped around the abdomen. This device helps
dividuals in whom a breathhold cannot be success- minimize respiratory excursions and minimizes the
fully maintained. positional variations of structures caused by breathing
The half-Fourier single-shot turbo spin echo motion. Finally, more sophisticated techniques with
(HASTE) sequence provides rapid breathhold-in- biofeedback strategies and navigator pulses are being
dependent T2-weighted imaging of the abdomen, investigated but are not readily available in the
allowing for a better success rate in imaging uncoop- clinical environment.
erative, somnolent, or poor breathholding patients. An elaborate technique used to suppress respira-
HASTE (Siemens Medical Solutions, Erlangen, Ger- tory motion in coronary imaging uses respiratory
many) or single-shot fast spin echo (SSFSE, GE gating with MR imaging navigators [75 – 77]. This
Medical Systems, Milwaukee, WI) are preferred technique has not yet been widely used for abdominal
as the motion-insensitive sequence for obtaining T2- imaging [78], although a recent report implementing
weighted images. navigators for renal artery imaging suggests its po-
The use of fat-suppression can augment image tential [79].
contrast and reduce artifacts caused by respiration
and other bulk motions. Many fat suppression strate- Role of coils and parallel imaging
gies increase the acquisition time but when applied
to motion insensitive nonbreathhold techniques can The use of localized coils (ie, phased array torso
offer improved image contrast and can identify the coil) is important to augment the SNR especially
presence of fat within a lesion. The authors have when considering the high bandwidth sequences
relied on fat suppression with HASTE and SSFSE for necessary for contemporary rapid imaging. The use
improving the image contrast with T2-weighted of systems with high performance gradients and new
sequences. To their knowledge there has not been a sequence designs has approached the limits on imag-
successful implementation of fat suppression to Mag- ing speed based on patient safety considerations. The
PrepGRE imaging. fastest techniques can result in peripheral nerve
stimulation, related to the maximum switching rates
Motion compensation strategies of magnetic field gradients.
A variety of means can be used to compensate for Parallel imaging or partially parallel imaging
in-plane motion artifacts resulting from respiration. techniques, such as sensitivity encoding, simulta-
These techniques typically add considerable time to neous acquisition of spatial harmonics, and array
the acquisition. Averaging strategies have been com- spatial sensitivity encoding technique, can be used
monly used to compensate for respiratory motion. to accelerate fast imaging sequences without increas-
The disadvantages of this technique include increased ing gradient switching rates or radiofrequency (RF)
scan time and obscured details. Image degradation is power deposition. This has been achieved by exploit-
most severe when breathing is erratic. ing spatial information inherent in the geometry of a
Perhaps more beneficial in those patients with surface coil array [80,81]. The coil profiles are used
erratic breathing patterns is the use of intermittent to generate missing k-space lines. This allows multi-
sampling of data at a relatively quiet time in the respi- ple lines in k space to be generated simultaneously
ratory cycle. This can be accomplished with respi- for application of each phase-encoding gradient.
884 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

The recent emergence of clinically approved T2-weighted imaging is most helpful in distinguish-
whole-body 3-T systems makes the use of the body ing simple cysts from other lesions. Simple cysts
coil without phased array coils feasible because the have much longer relaxation times than renal paren-
SNR is twice as high at 3 T as at 1.5 T. High field chyma and readily detected as high signal intensity
strength also extends the ultimate capabilities of lesions on T2-weighted imaging techniques. Septa-
parallel imaging. Acceleration in parallel MR imag- tions can be depicted readily within cysts (Fig. 4) and
ing comes at a cost in SNR: the higher the accelera- those cysts complicated by hemorrhage or infection
tion, the greater the SNR penalty. may be heterogeneous or low in SI on T2-weighted
The inherent SNR advantage at 3 T (resulting images. Angiomyolipoma (AML), hematoma, aneu-
from increased spin polarization) allows for greater rysm, and infectious mass can all demonstrate het-
acceleration factors or higher degrees of resolution to erogeneous T2 signal intensities.
be pursued while maintaining image quality. More- Renal cell carcinoma is variable in signal on
over, recent theoretical investigations [82,83] at the T2-weighted images [84]. Hemorrhagic products in
authors’ center and elsewhere have predicted that the malignant tumors cause heterogeneous T2 signal
combination of high field strength and parallel imag- characteristics, but cannot be distinguished reliably
ing will afford SNR advantages above and beyond from benign cysts containing hemorrhage. Defini-
those resulting from increased spin polarization. In tive characterization depends on the demonstration
particular, the increased ability to focus radiofre- of enhancement within a lesion to identify a vascular
quency energy at high RF frequencies has been supply; that demonstration excludes a simple cyst
shown to result in an improved capacity for spatial (Fig. 5).
encoding with coil arrays, and hence in higher SNR
and higher achievable accelerations for high-field Spin echo sequences
parallel imaging.
In conventional spin echo imaging only one phase-
encoding step per TR is used to encode spatial infor-
T2-weighted imaging mation. The associated acquisition times exceed the
possibility for breathhold imaging. These sequences
Clinical applications clearly benefit from the previously mentioned strate-
gies of suppressing motion-induced artifacts. Regard-
Normal kidney has a relatively long T2 time, less, motion-induced blurring and ghost artifacts
yielding a higher SI relative to liver and many other remain a problem. Fat suppression techniques can
soft tissues but close in SI to that of the spleen. augment image contrast and reduce motion arti-

Fig. 4. Renal cyst with multiple septations in 88-year-old man with cystic lesion in the right kidney on prior abdominal CT
performed for staging of bladder carcinoma. (A) Cor T2. Coronal fat saturated HASTE image (TR = 1100, TE = 64, FA = 130,
slice thickness = 4 mm) of the abdomen shows a large cyst in the right kidney with multiple septations (arrow). A smaller cyst is
noted in the medial aspect of the upper pole of the right kidney (arrowhead). (B) Cor T2 subs 2. Coronal three-dimensional fat-
saturated T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) after
administration of gadolinium demonstrates the lack of enhancement confirming the cystic nature of the lesion. A thin septation is
noted in the superior aspect of the cyst (arrowhead). A second small nonenhancing cyst also is noted in the upper pole of the
right kidney (arrow).
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 885

Fig. 5. Coexistent papillary renal cell carcinoma and hemorrhagic cyst in a 65-year-old man with esophageal carcinoma. Renal
mass incidentally noted on CT scan. (A) Coronal HASTE image (TR = 1100, TE = 64, FA = 130, slice thickness = 4 mm) at the
level of the kidneys shows two lesions in the right kidney, one in the superior (arrow) pole, and one in the inferior (arrowhead)
pole. The lesion in the upper pole is slightly hypointense compared with the renal parenchyma. The lesion in the inferior pole is
isointense to the renal parenchyma. Based on these findings, differentiation between cyst and solid tumor cannot be achieved.
(B) Unenhanced, three-dimensional fat-saturated T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice
thickness = 4 mm, before interpolation) shows the large lesion in the superior pole of the right kidney with similar signal
intensity than the renal medulla. This finding does not help in its characterization. Note that the lesion in the inferior pole
demonstrates high signal intensity suggesting hemorrhage. (C) Gadolinium-enhanced, three-dimensional fat-saturated
T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) during the
venous phase shows both lesions with signal intensity lower than that of the enhancing renal parenchyma. Determination of
enhancement of these lesions is difficult based on subjective impression alone. (D) Coronal subtracted three-dimensional fat-
saturated T1-weighted gradient echo image (venous phase [Fig. 5C] minus precontrast [Fig. 5B]) confirms the enhancement of
the lesion in the superior pole (arrow) of the right kidney. In contrast, the inferior pole lesion appears black (arrowhead)
because of the lack of enhancement. The lesions were confirmed to be a papillary RCC in the upper pole of the right kidney
and a hemorrhagic cyst in the inferior pole at pathology.

facts caused by respiration, but further increase ac- T2-weighted images in less time than conventional
quisition time. spin echo imaging. The length of the echo train is
proportional to the reduction in scan time that can be
Echo-train imaging achieved. Longer echo-trains can be used to obtain
T2-weighted images in the time frame of a breath-
Echo-train imaging is generically referred to as hold, which have been shown to improve results
‘‘rapid acquisition with relaxation enhancement [86,87]. Strong gradient systems benefit echo-train
sequences’’ [85]. These techniques commonly use imaging by minimizing interecho spacing, which in
vendor-related acronyms, such as fast spin echo turn reduces artifacts.
and turbo spin echo. This family of sequences is The effective echo time is determined by the echo
characterized by the application of a train of multiple times of the lowest phase-encoding gradients among
phase-encoding gradients for a given TR, obtaining the echo train (the center of k space) and provides the
886 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

dominant image contrast. Fat demonstrates higher T1-weighted imaging


signal intensity with echo-train techniques compared
with conventional T2-weighted SE sequences. Selec- Clinical applications
tion of the effective echo time and the addition of fat-
suppression, as needed, maximize image contrast In the normal kidney the cortex is slightly higher
with echo-train imaging. in signal intensity than the medulla on strongly
As mentioned previously, nonbreathhold, echo- T1-weighted sequences and the medulla has a similar
train, T2-weighted imaging can be improved by the signal intensity compared with muscle. Most cysts
use of fat suppression or respiratory triggering for have a long T1, and often appear lower in signal
patients with limited breathhold capacity. An alterna- intensity than normal renal parenchyma. Cysts com-
tive motion reduction strategy is the placement of a plicated by hemorrhage or containing proteinaceous
saturation band over the anterior abdominal wall fluid may demonstrate increased signal intensity
subcutaneous fat. This nullifies signal from the tissue because of T1 shortening effects from blood products
most responsible for propagating respiratory artifacts or protein and may show heterogeneous signal fea-
into the abdominal cavity. tures or fluid-fluid levels (Fig. 6).
Most of the solid renal masses other than AML
demonstrate signal intensity that is slightly lower
Half-Fourier T2 weighted imaging compared with renal cortex on T1-weighted images.
Macroscopic fat, found in most AMLs, has a short T1
Half-Fourier reconstruction is another modifica- and exhibits a relatively high signal on T1-weighted
tion that can be added to echo-train imaging further to images compared with background parenchyma. High
decrease the acquisition time (eg, HASTE, SSFSE). signal on T1-weighted images can also be caused by
HASTE is a single-shot technique that acquires just paramagnetic effects, such as intralesional hemorrhage
over half of k-space in one echo-train. The symmetry (cyst, AML, and RCC); melanin-containing lesions
of k space allows for mathematic reconstruction of an (metastases from malignant melanoma); and proteina-
image based on that partial acquisition [88]. ceous mucin-containing lesions (complicated cyst and
Because data can be acquired in less than a abscess). The presence of macroscopic fat is best
second on a per-slice basis, HASTE is very helpful confirmed with the use of fat-saturation techniques.
in patients unable to breathhold or when a rapid Some AMLs have less fatty component, and
survey is needed. Compared with turbo spin echo, may be difficult to be differentiated from other solid
HASTE can have a shorter TE because the center renal lesions. In this regard chemical-shift imaging
of k-space can be acquired near the start of the (in-phase and opposed-phase gradient echo [GRE]
echo-train [89]. imaging) is an important tool to detect microscopic,
Drawbacks of half-Fourier imaging include fractional intravoxel lipid, with certain caveats as
poorer SNRs than those found in turbo spin echo, described later in this manuscript.
and reduced contrast-to-noise ratio compared with
conventional T2-weighted spin echo or sequences GRE sequences
using shorter echo-trains. In addition, blurring is seen
as a result of T2 decay during the long echo-train. The GRE techniques for T1-weighted imaging
These render HASTE less sensitive for small, low- include either multishot or single-shot strategies.
contrast lesions. The most widely used and efficacious GRE technique
The authors use this technique to survey for the is the multishot, spoiled approach. Such sequences as
presence of focal lesions and as a rapid assessment fast spoiled GRE sequences (GE Medical Systems)
for hydronephrosis or collecting system filling de- and fast low-angle shot sequences (Siemens Medical
fects. In the authors’ experience HASTE techniques Systems), use short TR and TE values with a flip
can characterize cysts when there is uniform, mark- angle of 70 to 90 degrees and can provide full
edly hyperintense signal without mural nodularity coverage of the kidney in one 18- to 23-second
or complicated internal septations, in essence serving breathhold. These acquisitions offer good SNR, reg-
much as an ultrasound for assessment of renal le- ular section spacing, and minimize respiratory-related
sions. HASTE also offers a rapid survey for pa- artifacts. To facilitate the breathhold while preserving
thology elsewhere in the abdomen and renders SNR and optimal contrast, a TR of 120 to 200 mil-
excellent anatomic details. Coronal HASTE has liseconds is advised. The speed of these sequences
become a routine component for the authors’ renal allows for dynamic contrast-enhanced MR imaging
imaging and for upper abdominal imaging in general. with or without fat suppression [90,91].
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 887

Fig. 6. Fluid-fluid level in hemorrhagic cyst in 64-year-old man with prostate cancer. (A) Axial T1-weighted in-phase gradient
echo image (TR = 160, TE = 5.3, FA = 90, slice thickness = 8 mm) at the level of the kidneys shows a right renal lesion with a
fluid-fluid level (arrow). Note the hyperintense blood products layering in the dependent portion of the lesion. (B) Contrast-
enhanced three-dimensional fat-saturated T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm,
before interpolation) during the delayed venous phase shows homogenous enhancement of both kidneys. Hyperintense blood
products within the cyst can be interpreted erroneously as a focus of enhancement (arrow). (C) Axial subtracted (postcontrast
minus precontrast) three-dimensional fat-saturated T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thick-
ness = 4 mm, before interpolation) demonstrates lack of enhancement in the right renal cyst (arrow).

It is the comparison of precontrast and delayed postcontrast imaging but can also be diagnosed by
(nephrographic) postcontrast T1-weighted images that using a flow-sensitive GRE sequence to distinguish
is key to the detection and characterization of renal between flowing blood and tumor thrombus [94].
lesions (Fig. 7) [3,92,93]. Tumor extension into ve- MagPrepGRE imaging is vital for providing diag-
nous structures is often readily determined on delayed nostic-quality T1-weigthed images in patients with a
888 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

Fig. 7. Characterization of renal mass with precontrast and postcontrast T1-weighted images in 69-year-old woman with gross
hematuria and right flank pain. (A) Unenhanced, coronal three-dimensional fat-saturated T1-weighted gradient echo image (TR =
4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation). A large mass is noted in the inferior pole of the right kidney.
The hypointense center (arrow) suggests necrosis, whereas areas of high signal intensity in the dependent aspect of the mass
(arrowhead) are consistent with hemorrhage. (B) Coronal three-dimensional fat-saturated T1-weighted gradient echo image
(TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) during the arterial phase after administration of
gadolinium shows early enhancement of the peripheral rim of tumor (arrows). There is suggestion of enhancing septae in the
center of the lesion (arrowhead). (C) Coronal three-dimensional fat-saturated T1-weighted gradient echo image (VIBE) (TR =
4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) during the venous phase after administration of gadolinium
better demonstrates the irregular enhancing septae within the mass (arrow) and areas of necrosis. At nephrectomy, a clear cell
RCC with cystic and hemorrhagic degeneration was found.

limited breathhold capacity. The magnetization-pre- In-phase and opposed phase


pared sequence is structured so that data acquisition
occurs during the T1 recovery of tissues following a The GRE images can be obtained with specific TE
180-degree inversion pulse. The inversion pulse pro- values such that protons from fat and water are either
vides flexible image contrast [95]. When a section- in-phase or out-of-phase with one another [96]. When
selective 180-degree inversion pulse is used, only the fat and water are present within a voxel, a loss of
protons of the specific section are inverted. This signal intensity is noted when the opposed-phase
results in images in which the vessels are bright images are compared with the in-phase images.
and lesions are dark. The authors use a nonselective Compared with frequency-selected fat suppression,
inversion pulse, which, with an appropriate TI time, opposed-phase technique is sensitive for detecting
yields dark blood and excellent T1 contrast. With intracellular lipid. Renal and adrenal masses may
this technique, pulsation artifacts are eliminated and contain focal fat (angiomyolipomas and myelolipo-
enhancement of vessels or tumor thrombus within mas, respectively) or diffuse, intracellular lipid (clear
veins can be readily appreciated. cell renal carcinomas and adenomas, respectively)
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 889

Fig. 8. Microscopic and macroscopic fat in angiomyolipoma in 59-year-old woman with renal mass incidentally noted on
ultrasound. (A) Axial T1-weighted in-phase gradient echo image (TR = 160, TE = 5.3, FA = 90, slice thickness = 8 mm) shows a
slightly heterogeneous hyperintense mass in the left kidney (arrow). (B) Axial T1-weighted out-of-phase gradient echo image
(TR = 160, TE = 2.7, FA = 90, slice thickness = 8 mm) at the same level as Fig. 8A demonstrates decreased signal intensity
within near the entire mass. This finding is consistent with the presence of fractional intravoxel fat and water (microscopic fat).
Central hyperintense areas that do not decrease in signal intensity (arrowhead) are likely caused by a focus of macroscopic fat.
(C) Unenhanced, coronal three-dimensional fat-saturated T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice
thickness = 4 mm, before interpolation) shows the mass in the left kidney with intermediate signal intensity. Fat saturation is
achieved by applying a frequency selective pulse. Note focal areas of low signal intensity consistent with saturation of the
macroscopic fat within the mass (arrow). The presence of macroscopic fat is virtually diagnostic of AML. (D) Coronal three-
dimensional fat-saturated T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before
interpolation) after administration of gadolinium demonstrates enhancement within the lesion confirming its solid and
heterogeneous nature (arrow).
890 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

[94,97,98]. AML may contain both macroscopic fat intensity on the opposed phase sequence is not
and intracellular lipid (Fig. 8). specific for angiomyolipoma [97,99]. Chemically
On opposed-phase images, some clear cell car- selective fat saturation techniques are primarily
cinomas show relative focal or diffuse loss of used for identifying masses containing macroscopic
signal intensity because of the presence of intracel- fat, such as AMLs and adrenal myelolipomas. On
lular fat (Fig. 9). In renal masses, loss of signal opposed-phased GRE images a boundary chemical

Fig. 9. Microscopic fat in renal cell carcinoma (RCC) in a 58-year-old man with right renal mass found on CT scan performed as
a work-up of varicocele. Histopathologic analysis after nephrectomy revealed RCC, clear cell type. (A) Axial T1-weighted
gradient echo in-phase image (TR = 160, TE = 5.3, FA = 90, slice thickness = 8 mm) demonstrates a large mass in the right
kidney. Note the heterogenous appearance with low signal intensity center (arrow) and a peripheral slightly hyperintense rim
(arrowheads). (B) Axial T1-weighted gradient echo out-of-phase image (TR = 160, TE = 2.7, FA = 90, slice thickness = 8 mm)
shows decreased signal in the peripheral rim (arrowheads), which now is isointense to the central area. This finding is consistent
with the presence of fractional intravoxel fat (microscopic fat). (C) Coronal three-dimensional fat-saturated T1-weighted gradient
echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) during the arterial phase after
administration of gadolinium confirms the solid nature of the mass with a thick peripheral rim of enhancing tumor (arrowheads)
and central necrosis (arrow).
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 891

shift artifact is frequently present surrounding the being slightly off the optimal TE values (eg, 5.3 milli-
fatty components of these masses, a finding that seconds and 2.7 milliseconds for in- and out-of-
can be helpful for characterizing these lesions phase, respectively), are still able to portray the
(Fig. 10). desired imaging features.
Perinephric involvement of tumor can also be The in-phase and opposed-phase images can be
detected using opposed-phase GRE images because acquired simultaneously in the multisection mode.
of the artificial accentuation of renal contours from This is accomplished by acquiring two echoes per
the black line at the renal-retroperitoneal fat inter- excitation at different echo times and reconstructing
face, established by chemical shift artifact [94]. In the data as separate image sets. Because this dual-
the authors’ experience, however, it is the negative echo, in-phase, opposed-phase sequence can be
predictive value, seen by the preservation of the obtained during a breathhold, respiratory misregistra-
black line between adjacent tissues, which has the tion between in-phase and opposed-phase images is
most value in excluding contiguous involvement of eliminated, which facilitates a comparison of the two
structures. When a mass extends beyond the renal image sets.
capsule and abuts an adjacent organ the black line A complete renal examination requires the use
may be lost without invasion of that adjacent organ. of T1-weighted images with chemically selective fat
At 1.5 T, a GRE sequence with a TE of 4.4 milli- saturation in addition to in- and opposed-phase
seconds yields an in-phase image, whereas a TE of imaging to capture the specificity of fat suppression
2.2 milliseconds yields an opposed-phase image. rather than relying on inferences to determine a
Some manufacturers use sequences that, despite mass containing bulk fat.

Fig. 10. Characterization of angiomyolipoma using chemical shift artifact in 57-year-old woman with hypertension. Left renal
lesion incidentally noted on MR angiography of the abdomen for evaluation of renal artery stenosis. (A) Axial T1-weighted
in-phase gradient echo image (TR = 160, TE = 5.3, FA = 90, slice thickness = 8 mm) shows a hyperintense lesion in the left
kidney (arrow). (B) Axial T1-weighted out-of-phase gradient echo image (TR = 160, TE = 2.7, FA = 90, slice thickness = 8 mm)
demonstrates a boundary chemical shift artifact at the interface of the lesion and the surrounding renal parenchyma caused by the
coexistence of intravoxel fat and water protons (arrow).
892 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

Spin echo sequences Three-dimensional Fourier transform imaging has


advantages over two-dimensional imaging. Properly
Formerly the standard for T1-weighted imaging structured three-dimensional GRE sequences, such as
of the body, spin echo sequences should be relegated volumetric interpolated breathhold examination (Sie-
to systems unable to achieve good-quality breathhold mens Medical Solutions) or fast acquisition with
imaging. These systems clearly benefit from the multiphase Efgre3d (GE Medical Systems), have
previously mentioned strategies aimed at suppressing the capacity to provide thin sections, no gaps, fat
motion-induced artifacts, but motion-induced blur- saturation, higher SNR ratios, and comparable image
ring of contours remains a problem. For spin echo contrast in a breathhold time frame [102].
T1-weighted imaging either a standard spin echo se- Breathhold contrast material enhanced three-
quence with a short TR (eg, 400 to 600 milliseconds) dimensional fast spoiled GRE sequences are crucial
or a short echo-train (eg, 3) turbo spin echo sequence for the evaluation of renal vascular structures [103].
with multiple signal averages is generally used, but The volumetric three-dimensional sequence, used for
breathhold imaging is difficult to accomplish. combined parenchymal and vascular imaging, is
The use of half-Fourier reconstructions with spin modified from sequences originally designed for
echo sequences can yield good-quality breathhold gadolinium-enhanced angiography [102]. Key attri-
images while minimizing susceptibility artifacts. butes of the sequence geared toward optimized
The rapid acquisition spin echo technique combines parenchymal evaluations include the use of reduced
a relatively short repetition time, a short echo time, flip angle (10 to 15 degrees) and a symmetric or full
and half-Fourier data sampling [100]. This spin echo echo for readout. When appropriately thin sections
technique is less vulnerable than GRE sequences to yield pixel sizes approaching nearly isotropic resolu-
susceptibility artifacts and can be beneficial following tion and accurate timing methods are used, a single
surgery with metallic clips. Rapid acquisition spin data set generates high-quality images of the paren-
echo generally requires two breathholds to achieve chyma as well as MR angiography (Fig. 11).
full anatomic coverage and suffers from relatively Furthermore, the thin sections that can be acquired
low SNRs. The latter can be largely overcome with with three-dimensional imaging yield a data set that is
the use of a phased-array body coil [101]. amenable to meaningful multiplanar reconstructions,
allowing images to be reformatted in standard or-
thogonal planes, oblique axes, and curved planes.
Two- versus three-dimensional T1-weighted spoiled This can facilitate characterization of lesions that may
GRE sequences be difficult to evaluate on axial images. Multiplanar
reconstructions of volumetric data are also useful for
Fast T1-weighted imaging with GRE sequences pretherapeutic planning including surgery, emboliza-
is the cornerstone of renal imaging. For two-dimen- tion, radiofrequency ablation, and cryoablation.
sional breathhold imaging in the abdomen, the quality A useful nuance that benefits three-dimensional
and efficacy of imaging are limited by the need to coronal acquisitions and coronal acquisitions in gen-
acquire enough sections to cover a relatively large eral is the placement of the patient’s arms outside the
region, typically 160 to 200 mm. This must be accom- imaged field of view. This avoids aliasing artifacts
plished with the finite number of sections that can be and allows for the smallest possible field of view and
obtained in less than 25 seconds and requires the use the highest resolution in a given acquisition time. The
of relatively thick sections (8 to 10 mm) and interslice patient’s arms can be elevated with towels or cush-
gaps. Alternatively, two separate image sets have to be ions. Alternatively, the patient’s arms can be raised
obtained to achieve adequate anatomic coverage. over their head.
With two-dimensional GRE imaging, smaller
lesions can be missed or insufficiently characterized
because of partial volume averaging and low con- Technical considerations for three-dimensional MR
trast-to-noise ratio. The short acquisition times may angiography
also place other serious trade-offs on imaging, such Dynamic coronal three-dimensional imaging is
as increasing anatomic coverage at the expense of the fundamental technique for MR angiography of
decreased spatial resolution, or requiring the use of the renal arteries. An excellent approach uses high
higher bandwidths that reduce the SNR. In addition, spatial resolution, small field of view, axial three-
some fat-saturation methods, which can improve dimensional contrast-enhanced MR angiography of
contrast-to-noise ratios on contrast-enhanced images, the renal arteries followed several minutes later by the
require additional imaging time. more standard large field of view, three-dimensional,
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 893

Fig. 11. Seventy-year-old man with incidental renal mass on CT scan. (A) Coronal three-dimensional fat-saturated T1-weighted
gradient echo image in the venous phase after administration of gadolinium. The use of a low flip angle and full echo allows for
a good visualization of the renal parenchyma. A large enhancing mass is visualized in the inferior pole of the left kidney (arrow).
(B) Coronal source image from the same acquisition as Fig. 11A at a slightly anterior location. Note the excellent visualization of
the enhancing normal left renal vein (arrow). (C) Maximum intensity projection from the subtracted coronal data set acquired
during the arterial phase. There is excellent visualization of both renal arteries (arrows) using the technique tailored for
evaluation of both vascular and parenchymal details.

coronal contrast-enhanced MR angiography, the latter typically referred to as ‘‘k space.’’ K space, or Fourier
including the distal aorta and iliac arteries [104]. space, does not map to the image pixel by pixel.
With dynamic three-dimensional Fourier imaging, Rather, the information within k space reflects the
the order in which data are acquired has a substantial spatial frequency features of the image. The low spa-
impact in imaging considerations [105,106]. The tial frequency information, in the center of k space,
matrix into which data are filled in MR imaging is dominates image contrast, whereas the higher spatial
894 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

frequency data, at the periphery of k space, determine ing the severity of disease. For assessing the renal
image detail [107]. To obtain an arterial-phase image arteries they are usually reserved for detection of
in which arteries are bright and veins are dark, it is main renal artery stenoses and as a noninvasive func-
essential that the central k-space data (ie, the low tional assessment.
spatial frequency data) are acquired while the gado-
linium concentration in the arteries is high but rela- Time of flight
tively lower in the veins.
The standard mode of filling k space is a se- Time of flight was formerly a standard technique
quential mode in which the most negative spatial for vascular imaging in the body. Although stenoses
frequency component is collected first, with each of the proximal renal arteries can be detected with
subsequent point advancing through the zero spatial this technique, accessory renal arteries of small cali-
frequency component and continuing to the most ber and distal branches of renal arteries are not
positive spatial frequency component. With centric adequately displayed [110]. Other disadvantages of
acquisitions, the zero spatial frequency component this technique compared with gadolinium-enhanced
in the xy plane is acquired first, followed by pro- three-dimensional MR angiography include degrada-
gressively higher spatial frequency components, tion from in-plane saturation and motion related
alternating between positive and negative. artifacts. Gadolinium-enhanced breathhold three-
Elliptic-centric filling of k space is an extension of dimensional technique improves the evaluation of
centric ordering. It first fills the center of k space in vascular anatomy and is more reliable for visual-
the xy and z planes (in-plane and through-plane axes, izing accessory renal arteries [24,111]. The emer-
respectively). In so doing the center portion of k space gence of gadolinium-enhanced three-dimensional
is effectively compressed and filled in a very short MR angiography has virtually eliminated non –
time. This approach has been used effectively for contrast-enhanced time of flight for abdominal MR
renal MR angiography [104]. angiography. For renal imaging time of flight can be
If the center of k space is inadvertently acquired helpful to clarify subtle intraluminal filling defects
too early, severe artifacts can result, particularly with within the veins where flow artifacts still can affect
centric acquisitions [105,108]. These artifacts are gadolinium MR angiography. For this purpose ECG-
manifested as ringing and widening of the apparent triggered cine sequences are particularly valuable.
lumen. Centric acquisitions, however, offer some
advantages. These techniques are more robust to Phase contrast
breathing artifacts in poor breathholders because the
motion during the center of k space has the greatest Phase-contrast MR imaging is a flow-based tech-
impact [109]; patients who can only hold their breath nique exploiting the relationship between moving
for a short time (ie, 6 seconds) have less breathing protons and their response to the application of
artifact in centrically ordered three-dimensional gradients. It has not been widely accepted as a
acquisitions as compared with linear ordered three- technique for generating MR angiography in the
dimensional acquisitions. Furthermore, an earlier abdomen, primarily because of lengthy acquisition
acquisition of the center of k space minimizes paren- times, artifacts, and difficulty in selecting the proper
chymal enhancement, improving the delineation of velocity-encoding gradient. Three-dimensional phase
more distant renal vessels in the hilum and early contrast has served a useful function as an adjunct in
intraparenchymal branches. MR angiography studies. Proton dephasing and the re-
sultant loss in signal intensity can be seen in the pres-
ence of hemodynamically significant stenoses [36].
Flow-sensitive imaging The three-dimensional phase-contrast MR angiog-
raphy combined with renal gadolinium MR angiog-
The intrinsic sensitivity of MR imaging to flowing raphy can decrease the number of false-positive
spins affords the visualization of vessels, and an interpretations [43,112,113].
assessment of flow both qualitatively and quantita- The phase-contrast sequence has also been used
tively. Flow-sensitive imaging can be used to com- for quantitative measurement of renal arterial flow in
plement the morphologic images of contrast-enhanced evaluation of renal artery stenosis [44,114]. Velocity
MR angiography by providing hemodynamic in- waveforms in the renal artery, akin to Doppler sono-
formation [37]. Compared with three-dimensional graphic tracings, can be measured noninvasively
contrast-enhanced MR angiography, however, flow- using phase-contrast velocity-encoded MR imaging
sensitive techniques are more prone to overestimat- [115,116]. The combined approach of three-dimen-
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 895

sional gadolinium MR angiography and cine PC flow Turbulent or nonlaminar flow, which may be
measurement was reported to reveal excellent inter- caused by tortuous vessels or abrupt change of lumi-
observer variability and almost perfect intermodality nal diameter, can lead to the dissipation of phase
agreement with digital subtraction angiography coherence, which manifests as signal loss on all
[26,117]. MR imaging techniques including phase contrast
With faster acquisition times, several phase-con- [110,118]. This may cause false-positive findings but
trast velocity measurements may be obtained in the can often be clarified by using gadolinium-enhanced
same setting as routine MR angiography. The advan- three-dimensional MR angiography [34,115].
tages of MR phase-contrast technique over sono-
graphic approaches are it is not limited by patient
body habitus, it is not adversely impacted by the Contrast-enhanced dynamic imaging
presence of bowel gas, and the location and angle of
velocity measurements using MR imaging technique Clinical application
are also more easily achieved. The disadvantages
include the lower temporal resolution compared with The presence of contrast enhancement is the most
Doppler sonography, the lower spatial resolution crucial criterion for distinguishing solid renal lesions
precluding the evaluation of small intrarenal vessels from cysts. Image acquisition in various vascular
[110,113], and reliance on consistent cardiac rhythms phases after intravenous administration of a single
for ECG triggering [115]. bolus of contrast material refines the diagnostic

Fig. 12. Multilocular cystic renal cell carcinoma (RCC) in a 41-year-old man with left renal mass incidentally noted on work-up
of right upper quadrant discomfort. (A) Coronal fat-saturated HASTE image (TR = 1100, TE = 64, FA = 130, slice thickness =
4 mm) at the level of the kidneys. A large cystic lesion is noted in the inferior pole of the left kidney. Note multiple thick
septations within the lesion (arrow). (B) Coronal subtracted three-dimensional fat-saturated T1-weighted gradient echo image
(venous phase, unenhanced). The enhancing septae are readily seen. Note an enhancing nodule at the confluence of multiple
septations (arrow). (C) Coronal subtracted three-dimensional fat-saturated T1-weighted gradient echo image (venous phase,
unenhanced) at a slightly different level than Fig. 12B. Multiple irregular enhancing septae are visualized within the lesion
(arrows). Histopathologic analysis was diagnostic for multilocular cystic RCC.
896 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

work-up of renal masses. Dynamic contrast-enhanced acquire only two data sets, the first timed for the
studies usually allow the detection of even small arterial phase and the second initiated 35 seconds
enhancing solid areas within a cystic renal mass after the first.
(Fig. 12) [94]. With use of fast GRE sequences, dynamic con-
The scanning protocol should include unenhanced trast-enhanced MR imaging with or without fat sup-
MR imaging followed by imaging during the arterial, pression has been used to characterize renal lesions
venous, and nephrographic phases. The arterial phase by means of a qualitative or quantitative analysis of
is the most important for depicting arterial anatomy signal intensity changes over time [90,91]. Qualita-
and in identifying hypertrophied columns of Bertin as tive assessment of enhancement works quite well
pseudotumors (Fig. 13). provided that the same sequences are used for pre-
The venous phase is essential for imaging the contrast and postcontrast imaging [6,14].
renal veins for possible tumoral extension and the The determination of subtle enhancement can
parenchymal organs for potential metastases, whereas benefit from a quantitative approach or subtraction
the nephrographic phase is the most sensitive for imaging [14,119]. Care should be exercised when
tumoral detection. The authors perform three-dimen- using quantitative criteria for characterization. There
sional GRE T1-weighted sequences in a multiphase is no standardized scale for signal intensity values
series. The first acquisition is timed to capture the and the degree of precontrast and postcontrast
arterial phase; a second acquisition is initiated 20 sec- changes varies among sequences. For quantitative
onds after the first and a third acquisition is initiated evaluations it is important to ensure that unen-
30 seconds after the previous. Alternatively, one can hanced and enhanced sequences are identical, includ-

Fig. 13. Pseudotumor in a 36-year-old man with right upper quadrant discomfort. (A) Left sagittal ultrasonographic image shows a
hypoechoic mass in the mid portion of the kidney (arrow). (B) Coronal three-dimensional fat-saturated T1-weighted gradient echo
image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) during the arterial phase after administration of
gadolinium. There is normal enhancement of the left kidney. There is normal corticomedullary enhancement in the region of
the suspected mass (arrow). (C) Coronal image from a dynamic data set using scanning parameters from Fig. 13B acquired
30 seconds after the arterial phase confirms the normal enhancement of the pseudomass (arrow), which continues to track with the
cortex and medulla. (D) Coronal image from a dynamic data set using scanning parameters from Fig. 13B acquired 90 seconds
after the arterial phase confirms the normal enhancement of the pseudomass (arrow), which follows normal renal parenchyma.
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 897

ing the use of matched receiver gain and attenua- contrast enhancement does not always indicate
tion values. malignancy. A host of solid benign renal tumors
Renal cell carcinoma is the most common ma- and inflammatory masses also enhance. Of these,
lignancy of the kidneys. It is typically hypovascular AML and oncocytomas are the most common.
compared with the renal cortex but on occasion can AML has an early peak enhancement, although
be hypervascular. Other malignancies, such as TCC, these tumors remain hypointense compared with
metastatic lesions, and renal lymphoma also en- renal cortex, followed by a subsequent vascular
hance. It is important to note that the presence of washout [90].

Fig. 14. Metastatic transitional cell carcinoma (TCC) in a 53-year-old man with painless hematuria. (A) Coronal HASTE image at
the level of the kidneys shows severe hydronephrosis of the right kidney and target-appearing metastatic lesions to the liver
(arrows). No renal mass is detected based on this image. (B) Coronal three-dimensional fat-saturated T1-weighted gradient echo
(TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) image during the arterial phase after contrast
administration in this patient is vital for demonstrating the presence of an infiltrative heterogeneous enhancing mass in the upper
pole of the right kidney (arrows). The extension of the mass along the collecting system and the preservation of the renal
morphology suggest the diagnosis of TCC. The liver metastases are again noted. (C) Coronal three-dimensional fat-saturated
T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) image during the
delayed phase after administration of gadolinium. The tumor is now less conspicuous because it is near isointense with the renal
parenchyma (arrows). A nodular feature of the tumor is now seen at the wall of the superior calyx, however, after being filled
with contrast (arrowhead). Multiphasic imaging allows for detection and characterization of lesions.
898 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

One of the most important applications for con- Timing of image acquisition is especially important
trast-enhanced dynamic imaging is three-dimensional for efficacious venous-free renal MR angiography.
contrast-enhanced MR angiography. Its application The use of fixed image delays can suffice in most
has made MR angiography a widely used modal- patients but is not recommended because it is not
ity for noninvasive evaluation of renovascular dis- tailored to each patient’s circulatory dynamics and
ease. Timing of image acquisition is often performed results in as high as a 20% failure rate in capturing
by a test bolus [120,121]. Extremely fast acquisi- the arterial phase [127].
tion sequences allow time-resolved contrast-en- Gradient echo imaging techniques are the corner-
hanced MR angiography for evaluation of renal stone for all timing strategies whether this be a test
artery stenosis without the application of a timing dose; fluoroscopic monitoring (eg, CAREBOLUS,
run [122 – 124]. Siemens Medical Systems); or automated detection
of the bolus (eg, SMARTPREP, GE Medical Sys-
Technical specifics tems) [128]. A timing bolus of as little as 0.5 to 1 mL
gadolinium can be injected [120,126], but 2 mL
Contrast media and volume gadolinium is the authors’ routine. The imaging delay
Gadopentetate dimeglumine (Magnevist, Berlex is determined in each patient on the basis of the
Laboratories, Wayne, NJ) at the dose of 0.1 mmol/kg results of the timing examination and has been
is routinely used at the authors’ institution, although described elsewhere [120].
no efficacy difference among the Food and Drug
Administration – approved extracellular gadolinium- Power injector use
chelate contrast agents has been demonstrated. This The routine use of MR imaging – compatible
dose corresponds to a volume of 0.2 mL/kg. If the power injectors is recommended to yield reliable and
patient’s weight is unknown, a contrast volume of reproducible contrast delivery for optimized results.
20 mL can be used for an adult patient. Another
contrast agent, gadobenate dimeglumine (Multi-
Hance, Bracco, Milan, Italy) is still in the clinical Image processing
evaluation phase. It has been shown to produce
higher SNR at when compared with gadopentetate For quantitative and semiquantitative image analy-
dimeglumine at the same dose of 0.1 mmol/kg [125]; sis, image postprocessing is often a routine part of
the possible impact on diagnostic efficacy awaits an MR imaging examination. This can be performed
further study. on a workstation and the processed images then
transferred to a picture archiving and communica-
Contrast rate tions system for display or printed out as hardcopies
A contrast rate of 2 mL/second produces excellent as needed. To maintain throughput, the image sets
dynamic contrast-enhanced images. A recent report obtained during an MR imaging examination are
showed no significant difference in image quality for transferred to independent workstations and pro-
renal MR angiography when 0.1 mmol/kg dosing was cessed with commercially available hardware and
compared at rates of 2 and 3 mL/second [126]. software. At the authors’ institution, this is performed
using the Advantage Windows workstation (GE
Flush volume and rate Medical Systems) or the Virtuoso workstation (Sie-
A flush with saline of 20 mL at the rate of 2 mL/ mens Medical Solutions).
second is recommended, although when intravenous
access is established in the hand, the authors recom- Subtraction for accurate detection of enhancement
mend using a 30-mL saline flush. The larger volume
is used to maximize the delivery of the contrast dose Subtraction of the nonenhanced image set from
to the heart, which ultimately determines the profile the contrast-enhanced data sets is often performed for
of contrast enhancement in the individual’s tissues. better detection of subtle enhancement and to im-
prove the image contrast between vessels and back-
Timing ground tissue. Subtraction is particularly helpful for
For dynamic contrast-enhanced imaging, precise detection of a small enhancing component within a
timing of image acquisition during selected periods of cystic renal lesion (Fig. 15). Each phase from the
enhancement (eg, renal arterial versus venous phase) dynamic contrast-enhanced MR imaging study is
can be accomplished. This precision can be vital both used as a template from which the unenhanced data
to lesion detection and characterization (Fig. 14). set is subtracted.
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 899

Fig. 15. Value of subtracted images for detection of subtle enhancement (same patient as Fig. 11). (A) Sagittal three-dimensional
fat-saturated T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before interpolation) shows
predominantly hyperintense signal intensity within the mass consistent with hemorrhagic products. Note a nodular hypointense
appearance within the anterior aspect of the lesion (arrowheads) and a nodule in the posterior aspect (arrow). (B) Sagittal three-
dimensional fat-saturated T1-weighted gradient echo image (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before
interpolation) at the same level as Fig. 15B after administration of gadolinium. The signal intensity of the hemorrhagic
component of the mass has dropped relatively to the enhanced kidney suggesting lack of enhancement. The anterior
(arrowheads) and posterior (arrow) nodular components of the mass seem to enhance. Subjective assessment, however, of subtle
enhancement in complex lesions can be difficult. (C) Sagittal image obtained after subtraction of precontrast image (Fig. 10B)
from postcontrast image (Fig. 10C). Enhancement of the anterior (arrowheads) and posterior (arrow) nodular component is now
readily seen. The signal intensity of the hemorrhagic component of the lesion appears black because of lack of enhancement.
900 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

Subtraction often serves as the first step for further ghosting artifact around the renal contour as an index
imaging processing, such as the three-dimensional of the degree of misregistration (Fig. 16).
reconstructions for MR angiography, including MIP
and VR [52]. Image subtraction improves the quality Three-dimensional reconstruction algorithms for
of renal MR angiography in terms of both contrast-to- vascular anatomy and surgical planning
noise ratio and visualization of the distal renal arteries
[129]. When using subtraction for the qualitative The volumetric data acquired by MR angiography
assessment of renal lesion enhancement, it is impor- provide visualization in arbitrary oblique planes of
tant to evaluate the degree of misregistration that may the renal arteries, allowing evaluation of stenosis
be present. The authors use the thickness of a that is unattainable with projection techniques used

Fig. 16. Pseudoenhancement caused by respiratory misregistration. (A) Coronal HASTE image at the level of the kidneys shows
a homogeneously hyperintense lesion in the inferior pole of the right kidney suggesting a simple cyst (arrow). (B) Coronal three-
dimensional fat-saturated T1-weighted gradient echo sequence (TR = 4.5 TE = 1.9, FA = 12, slice thickness = 4 mm, before
interpolation) after administration of gadolinium confirms the presence of a nonenhancing simple cyst in the inferior pole of the
right kidney (arrow). (C) Coronal subtracted image (venous phase, unenhanced) shows a focus of high signal intensity in the
inferior pole of the cyst (arrow). This can be misinterpreted as enhancement. Note the rim of hyperintensity (ghosting artifact)
around the superior pole of both kidneys (arrowheads) related to respiratory misregistration between the precontrast and
postcontrast images. Careful review of source images and, if needed, a quantitative comparison of SI between unenhanced and
enhanced images are important for identifying pseudoenhancement.
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 901

Fig. 17. Retroaortic renal vein in 59-year-old man with painless hematuria. (A) Coronal subtracted three-dimensional fat-
saturated T1-weighted gradient echo image (venous phase, unenhanced) demonstrates enhancement of a lesion at the lower pole
of the left kidney (arrow). (B) Coronal volume-rendered image obtained from the same postcontrast phase as in Fig. 17A. There
are two renal arteries for the left kidney (arrowheads) and a retroaortic renal vein (arrow) is readily noted. Information about the
vascular anatomy is important before nephrectomy, particularly when laparoscopic approach is to be attempted.

in conventional angiography [19]. Because source The use of multiplanar reformations affords
images are still subject to partial volume effects unique perspective and can improve diagnostic accu-
[130], three-dimensional reconstructions have been racy [137,138]. Other reconstruction techniques, such
performed to enhance diagnostic confidence, to de- as shaded-surface display and virtual intra-arterial
lineate better the vascular anatomy, and to define endoscopy, seem to be time-consuming without pro-
better its relationship to adjacent structures [52]. viding much useful diagnostic gain [137]. One inter-
The MIP algorithm is the most widely used post- esting technical development for processing image
processing technique for MR angiography and the data attempts to quantify the degree of renal artery
resulting images are readily accepted by referring stenosis automatically by using a computer system
physicians. As the first step of reconstruction, the non- [139]. This approach has potential to provide rapid and
enhanced data set is subtracted from that of the arterial reproducible results but requires clinical validation.
phase. Targeted MIP images are then obtained in Both VR and multiplanar reformations have been
coronal and transverse projections selected to encom- used in therapeutic planning for total and partial
pass the renal artery from the origin to the renal hilum. nephrectomy surgery. The authors’ urologists have
Volume rendering images also are generally cre- found useful a preoperative review that offers multi-
ated in transverse and coronal orientations to demon- ple surgical perspectives. In essence with interactive
strate the renal artery from its origin to the renal VR and progressive restrictions on the data set
hilum. VR has been implemented as a three-dimen- presented, the resultant virtual surgery can reveal
sional reconstruction technique for postprocessing of the tissues and structures that are encountered during
renal CT angiography [131], and its application in the a particular surgical approach (Fig. 17)
evaluation of MR angiographic images has also been
investigated. It has been reported that VR algorithm
improves perceptibility and precision in the evalua-
tion of renal arterial lumina when compared with MIP Summary
algorithm [132,133], with a higher specificity and
accuracy especially with regard to detection of mod- This article describes the principles, attributes,
erate and severe stenosis [52,132]; the VR algorithm and pitfalls of the many MR imaging approaches
also better delineates overlapping vascular structures available for assessment of renal-related disorders.
[52,134]. Further information regarding the principles Tables 1 and 2 summarize the specific approach
of VR can be obtained elsewhere [131,135,136]. and rationale.
902 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

References logic-pathologic correlation. Radiographics 1998;18:


123 – 42; quiz 148.
[1] Nelson KL, Gifford LM, Lauber-Huber C, Gross CA, [16] Neri E, Boraschi P, Caramella D, Battolla L, Gigoni
Lasser TA. Clinical safety of gadopentetate dimeglu- R, Armillotta N, et al. MR virtual endoscopy of the
mine. Radiology 1995;196:439 – 43. upper urinary tract. AJR Am J Roentgenol 2000;175:
[2] Prince MR, Arnoldus C, Frisoli JK. Nephrotoxicity of 1697 – 702.
high dose gadolinium compared with iodinated con- [17] Qanadli SD, Soulez G, Therasse E, Nicolet V, Turpin
trast. J Magn Reson Imaging 1996;6:162 – 6. S, Froment D, et al. Detection of renal artery stenosis:
[3] Rofsky NM, Weinreb JC, Bosniak MA, Libes RB, prospective comparison of captopril-enhanced Dopp-
Birnbaum BA. Renal lesion characterization with ler sonography, captopril-enhanced scintigraphy, and
gadolinium-enhanced MR imaging: efficacy and MR angiography. AJR Am J Roentgenol 2001;
safety in patients with renal insufficiency [see com- 177:1123 – 9.
ments]. Radiology 1991;180:85 – 9. [18] Schoenberg SO, Prince MR, Knopp MV, Allenberg
[4] Smith SJ, Bosniak MA, Megibow AJ, Hulnick DH, JR. Renal MR angiography. Magn Reson Imaging
Horii SC, Raghavendra BN. Renal cell carcinoma: Clin N Am 1998;6:351 – 70.
earlier discovery and increased detection [see com- [19] Dong Q, Schoenberg SO, Carlos RC, Neimatallah M,
ments]. Radiology 1989;170(3 pt 1):699 – 703. Cho KJ, Williams DM, et al. Diagnosis of renal vas-
[5] Andre M, Helenon O, De Fromont M, Correas J, cular disease with MR angiography. Radiographics
Petit P, Bartoli J, et al. Kidney tumors: clinical and 1999;19:1535 – 54.
pathological findings and detection. J Radiol 2002; [20] Vasbinder GB, Nelemans PJ, Kessels AG, Kroon AA,
83(6 pt 2):773 – 83. de Leeuw PW, van Engelshoven JM. Diagnostic tests
[6] Semelka RC, Shoenut JP, Kroeker MA, MacMahon for renal artery stenosis in patients suspected of hav-
RG, Greenberg HM. Renal lesions: controlled com- ing renovascular hypertension: a meta-analysis. Ann
parison between CT and 1.5-T MR imaging with non- Intern Med 2001;135:401 – 11.
enhanced and gadolinium-enhanced fat-suppressed [21] Knopp MV, Floemer F, Schoenberg SO, von Tengg-
spin-echo and breath-hold FLASH techniques [see Kobligk H, Bock M, van Kaick G. Non-invasive
comments]. Radiology 1992;182:425 – 30. assessment of renal artery stenosis: current concepts
[7] Tello R, Davison BD, O’Malley M, Fenlon H, Thom- and future directions in magnetic resonance angiog-
son KR, Witte DJ, et al. MR imaging of renal masses raphy. J Comput Assist Tomogr 1999;23(suppl 1):
interpreted on CT to be suspicious. AJR Am J Roent- S111 – 7.
genol 2000;174:1017 – 22. [22] Gilfeather M, Yoon HC, Siegelman ES, Axel L,
[8] Birnbaum BA, Maki DD, Chakraborty DP, Jacobs JE, Stolpen AH, Shlansky-Goldberg RD, et al. Renal
Babb JS. Renal cyst pseudoenhancement: evaluation artery stenosis: evaluation with conventional angiog-
with an anthropomorphic body CT phantom. Radiol- raphy versus gadolinium-enhanced MR angiography.
ogy 2002;225:83 – 90. Radiology 1999;210:367 – 72.
[9] Maki DD, Birnbaum BA, Chakraborty DP, Jacobs [23] Hany TF, Debatin JF, Leung DA, Pfammatter T. Eval-
JE, Carvalho BM, Herman GT. Renal cyst pseudo- uation of the aortoiliac and renal arteries: comparison
enhancement: beam-hardening effects on CT num- of breath-hold, contrast-enhanced, three-dimensional
bers. Radiology 1999;213:468 – 72. MR angiography with conventional catheter angiog-
[10] Kozlowski PM, Winfield HN. Laparoscopic partial raphy. Radiology 1997;204:357 – 62.
nephrectomy and wedge resection for the treat- [24] Bakker J, Beek FJ, Beutler JJ, Hene RJ, de Kort GA,
ment of renal malignancy. J Endourol 2001;15: de Lange EE, et al. Renal artery stenosis and acces-
369 – 74. sory renal arteries: accuracy of detection and visual-
[11] Rosin A, Bauer JJ, Swanson SJ, Spevak M, Costabile ization with gadolinium-enhanced breath-hold MR
RA. Changing trends in partial nephrectomy at Wal- angiography. Radiology 1998;207:497 – 504.
ter Reed Army Medical Center. Mil Med 2001;166: [25] Tan KT, van Beek EJ, Brown PW, van Delden OM,
416 – 8. Tijssen J, Ramsay LE. Magnetic resonance angiogra-
[12] Rendon RA, Stanietzky N, Panzarella T, Robinette M, phy for the diagnosis of renal artery stenosis: a meta-
Klotz LH, Thurston W, et al. The natural history of analysis. Clin Radiol 2002;57:617 – 24.
small renal masses. J Urol 2000;164:1143 – 7. [26] Schoenberg SO, Knopp MV, Londy F, Krishnan S,
[13] Bosniak MA, Birnbaum BA, Krinsky GA, Waisman Zuna I, Lang N, et al. Morphologic and functional
J. Small renal parenchymal neoplasms: further obser- magnetic resonance imaging of renal artery stenosis:
vations on growth [see comments]. Radiology 1995; a multireader tricenter study. J Am Soc Nephrol 2002;
197:589 – 97. 13:158 – 69.
[14] Rofsky NM, Bosniak MA. MR imaging in the eval- [27] Mittal TK, Evans C, Perkins T, Wood AM. Renal
uation of small (< or = 3.0 cm) renal masses. Magn arteriography using gadolinium enhanced 3D MR
Reson Imaging Clin N Am 1997;5:67 – 81. angiography: clinical experience with the technique,
[15] Wong-You-Cheong JJ, Wagner BJ, Davis Jr CJ. Tran- its limitations and pitfalls. Br J Radiol 2001;74:
sitional cell carcinoma of the urinary tract: radio- 495 – 502.
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 903

[28] Chan YL, Leung CB, Yu SC, Yeung DK, Li PK. [42] Schoenberg SO, Bock M, Aumann S, Just A, Essig
Comparison of non-breath-hold high resolution M, Floemer F, et al. Quantitative recording of renal
gadolinium-enhanced MRA with digital subtraction function with magnetic resonance tomography. Radi-
angiography in the evaluation on allograft renal ologe 2000;40:925 – 37.
artery stenosis. Clin Radiol 2001;56:127 – 32. [43] Marcos HB, Choyke PL. Magnetic resonance
[29] Korst MB, Joosten FB, Postma CT, Jager GJ, angiography of the kidney. Semin Nephrol 2000;
Krabbe JK, Barentsz JO. Accuracy of normal-dose 20:450 – 5.
contrast-enhanced MR angiography in assessing re- [44] Schoenberg SO, Essig M, Bock M, Hawighorst H,
nal artery stenosis and accessory renal arteries. AJR Sharafuddin M, Knopp MV. Comprehensive MR
Am J Roentgenol 2000;174:629 – 34. evaluation of renovascular disease in five breath
[30] De Cobelli F, Venturini M, Vanzulli A, Sironi S, holds. J Magn Reson Imaging 1999;10:347 – 56.
Salvioni M, Angeli E, et al. Renal arterial stenosis: [45] Grist TM. Magnetic resonance angiography of renal
prospective comparison of color Doppler US and arterial stenosis. Coron Artery Dis 1999;10:151 – 6.
breath-hold, three-dimensional, dynamic, gado- [46] Radermacher J, Weinkove R, Haller H. Techniques
linium-enhanced MR angiography. Radiology 2000; for predicting a favourable response to renal angio-
214:373 – 80. plasty in patients with renovascular disease. Curr
[31] Thornton MJ, Thornton F, O’Callaghan J, Varghese Opin Nephrol Hypertens 2001;10:799 – 805.
JC, O’Brien E, Walshe J, et al. Evaluation of dynamic [47] Cieszanowski A, Golebiowski M, Szeszkowski W,
gadolinium-enhanced breath-hold MR angiography Symonides B, Gaciong Z. Morphologic and func-
in the diagnosis of renal artery stenosis. AJR Am J tional assessment of renal artery stenosis: use of
Roentgenol 1999;173:1279 – 83. combined MR angiography and MR renography –
[32] Thornton J, O’Callaghan J, Walshe J, O’Brien E, preliminary report. Pol Arch Med Wewn 2001;105:
Varghese JC, Lee MJ. Comparison of digital subtrac- 461 – 7.
tion angiography with gadolinium-enhanced mag- [48] Ros PR, Gauger J, Stoupis C, Burton SS, Mao J,
netic resonance angiography in the diagnosis of Wilcox C, et al. Diagnosis of renal artery stenosis:
renal artery stenosis. Eur Radiol 1999;9:930 – 4. feasibility of combining MR angiography, MR renog-
[33] Hany TF, Leung DA, Pfammatter T, Debatin JF. Con- raphy, and gadopentetate-based measurements of
trast-enhanced magnetic resonance angiography of glomerular filtration rate. AJR Am J Roentgenol
the renal arteries: original investigation. Invest Radiol 1995;165:1447 – 51.
1998;33:653 – 9. [49] Grenier N, Trillaud H, Combe C, Degreze P, Jeandot
[34] Shetty AN, Bis KG, Vrachliotis TG, Kirsch M, Shir- R, Gosse P, et al. Diagnosis of renovascular hyper-
khoda A, Ellwood R. Contrast-enhanced 3D MRA tension: feasibility of captopril-sensitized dynamic
with centric ordering in k space: a preliminary clinical MR imaging and comparison with captopril scintig-
experience in imaging the abdominal aorta and renal raphy. AJR Am J Roentgenol 1996;166:835 – 43.
and peripheral arterial vasculature. J Magn Reson [50] Krause UJ, Pabst T, Kostler H, Helbig C, Kenn W,
Imaging 1998;8:603 – 15. Hahn D. Time-resolved MR angiography of the renal
[35] Tello R, Thomson KR, Witte D, Becker GJ, Tress artery: morphology and perfusion. Rofo Fortschr
BM. Standard dose Gd-DTPA dynamic MR of renal Geb Rontgenstr Neuen Bildgeb Verfahr 2002;174:
arteries. J Magn Reson Imaging 1998;8:421 – 6. 1170 – 4.
[36] Prince MR. Renal MR angiography: a comprehensive [51] Coulam CH, Bouley DM, Sommer FG. Measurement
approach. J Magn Reson Imaging 1998;8:511 – 6. of renal volumes with contrast-enhanced MRI.
[37] Leung DA, Hagspiel KD, Angle JF, Spinosa DJ, Mat- J Magn Reson Imaging 2002;15:174 – 9.
sumoto AH, Butty S. MR angiography of the renal [52] Mallouhi A, Schocke M, Judmaier W, Wolf C,
arteries. Radiol Clin North Am 2002;40:847 – 65. Dessl A, Czermak BV, et al. 3D MR angiography
[38] Verschuyl EJ, Kaatee R, Beek FJ, Patel NH, Fon- of renal arteries: comparison of volume rendering
taine AB, Daly CP, et al. Renal artery origins: best and maximum intensity projection algorithms. Radi-
angiographic projection angles. Radiology 1997;205: ology 2002;223:509 – 16.
115 – 20. [53] Vallee JP, Lazeyras F, Khan HG, Terrier F. Absolute
[39] Tello R, Chaoui A, Hymphrey M, Fenlon H, Anasta- renal blood flow quantification by dynamic MRI and
sia-Rubino L, DeCarvalho VL, et al. Incidence of adre- Gd-DTPA. Eur Radiol 2000;10:1245 – 52.
nal masses in patients referred for renal artery stenosis [54] Binkert CA, Hoffman U, Leung DA, Matter HG,
screening MR. Invest Radiol 2001;36:518 – 20. Schmidt M, Debatin JF. Characterization of renal
[40] Gill IS, Meraney AM, Bravo EL, Novick AC. Pheo- artery stenoses based on magnetic resonance renal
chromocytoma coexisting with renal artery lesions. flow and volume measurements. Kidney Int 1999;
J Urol 2000;164:296 – 301. 56:1846 – 54.
[41] Camberos A, Bautista N, Rubenzik M, Applebaum [55] Prasad PV, Priatna A. Functional imaging of the kid-
H. Renal artery stenosis and pheochromocytoma: neys with fast MRI techniques. Eur J Radiol 1999;29:
coexistence and treatment. J Pediatr Surg 2000;35: 133 – 48.
714 – 6. [56] Namimoto T, Yamashita Y, Mitsuzaki K, Nakayama
904 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

Y, Tang Y, Takahashi M. Measurement of the ap- in healthy and pulmonary-compromised populations:


parent diffusion coefficient in diffuse renal disease effects of hyperventilation and oxygen inspiration.
by diffusion-weighted echo-planar MR imaging. J Magn Reson Imaging 1997;7:595 – 7.
J Magn Reson Imaging 1999;9:832 – 7. [70] Feinberg D, Rofsky NM, Johnson G. Multiple
[57] Israel GM, Lee VS, Edye M, Krinsky GA, Lavelle breath-hold averaging (MBA) method for increased
MT, Diflo T, et al. Comprehensive MR imaging in the SNR in abdominal MRI. Magn Reson Med 1995;
preoperative evaluation of living donor candidates for 34:905 – 9.
laparoscopic nephrectomy: initial experience. Radiol- [71] Haase A, Matthaei D, Bartkowski R, Duhmke E, Leib-
ogy 2002;225:427 – 32. fritz D. Inversion recovery snapshot FLASH MR
[58] Huber A, Heuck A, Scheidler J, Holzknecht N, Baur imaging. J Comput Assist Tomogr 1989;13:1036 – 40.
A, Stangl M, et al. Contrast-enhanced MR angiogra- [72] Matthaei D, Haase A, Henrich D, Duhmke E. Fast
phy in patients after kidney transplantation. Eur Ra- inversion recovery T1 contrast and chemical shift
diol 2001;11:2488 – 95. contrast in high-resolution snapshot FLASH MR
[59] Luk SH, Chan JH, Kwan TH, Tsui WC, Cheung YK, images. Magn Reson Imaging 1992;10:1 – 6.
Yuen MK. Breath-hold 3D gadolinium-enhanced sub- [73] Bailes D, Gilderdale DJ, Bydder GM, Collins AG,
traction MRA in the detection of transplant renal Firmin DN. Respiratory ordered phase encoding
artery stenosis. Clin Radiol 1999;54:651 – 4. (ROPE): a method for reducing respiratory motion
[60] Cambria RP, Kaufman JL, Brewster DC, Gertler JP, artefacts in MR imaging. J Comput Assist Tomogr
LaMuraglia GM, Bazari H, et al. Surgical renal artery 1985;9:835 – 8.
reconstruction without contrast arteriography: the role [74] Lewis C, Prato FS, Drost DJ, Nicholson RL. Com-
of clinical profiling and magnetic resonance angiog- parison of respiratory triggering and gating tech-
raphy. J Vasc Surg 1999;29:1012 – 21. niques for the removal of respiratory artifacts in
[61] Ferreiros J, Mendez R, Jorquera M, Gallego J, Lezana MR imaging. Radiology 1986;160:803 – 10.
A, Prats D, et al. Using gadolinium-enhanced three- [75] McConnell MV, Khasgiwala VC, Savord BJ, Chen
dimensional MR angiography to assess arterial in- MH, Chuang ML, Edelman RR, et al. Prospective
flow stenosis after kidney transplantation. AJR Am adaptive navigator correction for breath-hold MR
J Roentgenol 1999;172:751 – 7. coronary angiography. Magn Reson Med 1997;37:
[62] Carlos RC, Prince MR, Ward JS, Stanley JC, Dong 148 – 52.
Q, Londy FJ. Renal anatomic changes on magnetic [76] Danias PG, McConnell MV, Khasgiwala VC, Chuang
resonance imaging and gadolinium-enhanced mag- ML, Edelman RR, Manning WJ. Prospective navi-
netic resonance angiography after renal revasculari- gator correction of image position for coronary MR
zation: original investigation. Invest Radiol 1998;33: angiography. Radiology 1997;203:733 – 6.
660 – 9. [77] Danias PG, Stuber M, Botnar RM, Kissinger KV,
[63] Engellau L, Olsrud J, Brockstedt S, Albrechtsson U, Edelman RR, Manning WJ. Relationship between
Norgren L, Stahlberg F, et al. MR evaluation ex vivo motion of coronary arteries and diaphragm during
and in vivo of a covered stent-graft for abdominal free breathing: lessons from real-time MR imaging.
aortic aneurysms: ferromagnetism, heating, artifacts, AJR Am J Roentgenol 1999;172:1061 – 5.
and velocity mapping. J Magn Reson Imaging 2000; [78] Foo TK, King KF. A computationally efficient
12:112 – 21. method for tracking reference position displace-
[64] Oto A, Herts BR, Remer EM, Novick AC. Inferior ments for motion compensation in magnetic reso-
vena cava tumor thrombus in renal cell carcinoma: nance imaging. Magn Reson Med 1999;42:548 – 53.
staging by MR imaging and impact on surgical treat- [79] Spuentrup E, Manning WJ, Bornert P, Kissinger KV,
ment. AJR Am J Roentgenol 1998;171:1619 – 24. Botnar RM, Stuber M. Renal arteries: navigator-gated
[65] Semelka RC, Shoenut JP, Magro CM, Kroeker MA, balanced fast field-echo projection MR angiography
MacMahon R, Greenberg HM. Renal cancer staging: with aortic spin labeling: initial experience. Radiol-
comparison of contrast-enhanced CT and gadolinium- ogy 2002;225:589 – 96.
enhanced fat-suppressed spin-echo and gradient- [80] Pruessmann KP, Weiger M, Scheidegger MB, Boe-
echo MR imaging. J Magn Reson Imaging 1993; siger P. SENSE: sensitivity encoding for fast MRI.
3:597 – 602. Magn Reson Med 1999;42:952 – 62.
[66] Ngaage DL, Sharpe DA, Prescott S, Kay PH. Safe [81] Sodickson DK, McKenzie CA. A generalized ap-
technique for removal of extensive renal cell tumors. proach to parallel magnetic resonance imaging. Med
Ann Thorac Surg 2001;71:1679 – 81. Phys 2001;28:1629 – 43.
[67] Stief CG, Schafers HJ, Kuczyk M, Anton P, Pethig K, [82] Ohliger M, Yeh E, McKenzie C, Sodickson D.
Truss MC, et al. Renal-cell carcinoma with intracaval Constraints on the performance of parallel magnetic
neoplastic extension: stratification and surgical tech- resonance imaging. Presented at the Scientific Meeting
nique. World J Urol 1995;13:166 – 70. of the International Society for Magnetic Resonance in
[68] Kramer LA. Magnetic resonance imaging of renal Medicine. Honolulu, May 18 – 24, 2002.
masses. World J Urol 1998;16:22 – 8. [83] Wiesinger F, Pruessmann K, Boesiger P. Inherent limi-
[69] Marks B, Mitchell DG, Simelaro JP. Breath-holding tation of the reduction factor in parallel imaging as a
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 905

function of field strength. Presented at the tenth Scien- [99] Outwater EK, Blasbalg R, Siegelman ES, Vala M. De-
tific Meeting of the International Society for Magnetic tection of lipid in abdominal tissues with opposed-
Resonance in Medicine. Honolulu, May 18 – 24, 2002. phase gradient-echo images at 1.5 T: techniques and
[84] Huch Boni RA, Krestin GP. Contrast-enhanced MR diagnostic importance. Radiographics 1998;18:
imaging of the kidneys and adrenal glands. Magn 1465 – 80.
Reson Imaging Clin N Am 1996;4:101 – 31. [100] Mirowitz S, Lee JK, Brown JJ, Eilenberg SS, Heiken
[85] Hennig J, Nauerth A, Friedburg H. RARE imaging: JP, Perman WH. Rapid acquisition spin-echo (RASE)
a fast imaging method for clinical MR. Magn Reson MR imaging: a new technique for reduction of
Med 1986;3:823 – 33. artifacts and acquisition time. Radiology 1990;175:
[86] Earls J, Rofsky NM, DeCorato DR, Krinsky GA, 131 – 5.
Weinreb JC. Echo-train STIR MRI of the liver: [101] Decorato DR, Rofsky NM, Earls JP, Krinsky GA,
comparison of breath-hold and non-breath-hold Weinreb JC. T-1 weighted sequences for hepatic
imaging strategies. J Magn Reson Imaging 1999; MRI: re-evaluation using a phased array coil. Clin
9:87 – 92. Imaging 1999;23:26 – 31.
[87] Gaa J, Hatabu H, Jenkins RL, Finn JP, Edelman RR. [102] Rofsky N, Lee VS, Laub G, Pollack MA, Krinsky
Liver masses: replacement of conventional T2- GA, Thomasson D, et al. Abdominal MR imaging
weighted spin-echo MR imaging with breath-hold with a volumetric interpolated breath-hold examina-
MR imaging. Radiology 1996;200:459 – 64. tion. Radiology 1999;212:876 – 84.
[88] Feinberg DA, Hale JD, Watts JC, Kaufman L, Mark [103] Heiss SG, Shifrin RY, Sommer FG. Contrast-
A. Halving MR imaging time by conjugation: dem- enhanced three-dimensional fast spoiled gradient-
onstration at 3.5 kG. Radiology 1986;161:527 – 31. echo renal MR imaging: evaluation of vascular
[89] Tang Y, Yamashita Y, Takahashi M. Ultrafast T2- and nonvascular disease. Radiographics 2000;20:
weighted imaging of the abdomen and pelvis: use 1341 – 52.
of single shot fast spin-echo imaging. J Magn Reson [104] Fain SB, King BF, Breen JF, Kruger DG, Riederer
Imaging 1998;8:384 – 90. SJ. High-spatial-resolution contrast-enhanced MR
[90] Scialpi M, Di Maggio A, Midiri M, Loperfido A, angiography of the renal arteries: a prospective com-
Angelelli G, Rotondo A. Small renal masses: assess- parison with digital subtraction angiography. Radiol-
ment of lesion characterization and vascularity on ogy 2001;218:481 – 90.
dynamic contrast-enhanced MR imaging with fat sup- [105] Maki JH, Prince MR, Londy FJ, Chenevert TL. The
pression. AJR Am J Roentgenol 2000;175:751 – 7. effects of time varying intravascular signal intensity
[91] Ho VB, Allen SF, Hood MN, Choyke PL. Renal and k-space acquisition order on three-dimensional
masses: quantitative assessment of enhancement MR angiography image quality. J Magn Reson Imag-
with dynamic MR imaging. Radiology 2002;224: ing 1996;6:642 – 51.
695 – 700. [106] Laub G. Principles of contrast-enhanced MR angiog-
[92] Yamashita Y, Miyazaki T, Hatanaka Y, Takahashi M. raphy: basic and clinical applications. Magn Reson
Dynamic MRI of small renal cell carcinoma. J Com- Imaging Clin N Am 1999;7:783 – 95.
put Assist Tomogr 1995;19:759 – 65. [107] Mezrich R. A perspective on K-space. Radiology
[93] Eilenberg SS, Lee JK, Brown J, Mirowitz SA, Tartar 1995;195:297 – 315.
VM. Renal masses: evaluation with gradient-echo [108] Lee VS, Martin DJ, Krinsky GA, Rofsky NM.
Gd-DTPA-enhanced dynamic MR imaging. Radiol- Gadolinium-enhanced MR angiography: artifacts
ogy 1990;176:333 – 8. and pitfalls. AJR Am J Roentgenol 2000;175:
[94] Krestin GP. Genitourinary MR: kidneys and adrenal 197 – 205.
glands. Eur Radiol 1999;9:1705 – 14. [109] Maki JH, Chenevert TL, Prince MR. The effects of
[95] Edelman R, Wallner B, Singer A, Atkinson DJ, Saini incomplete breath-holding on 3D MR image quality.
S. Segmented turboFLASH: method for breath-hold J Magn Reson Imaging 1997;7:1132 – 9.
MR imaging of the liver with flexible contrast. Radi- [110] Loubeyre P, Trolliet P, Cahen R, Grozel F, Labeeuw
ology 1990;177:515 – 21. M, Minh VA. MR angiography of renal artery steno-
[96] Wehrli F, Perkins TG, Shimakawa A, Roberts F. sis: value of the combination of three-dimensional
Chemical shift-induced amplitude modulations in time-of-flight and three-dimensional phase-contrast
images obtained with gradient refocusing. Magn MR angiography sequences. AJR Am J Roentgenol
Reson Imaging 1987;5:157 – 8. 1996;167:489 – 94.
[97] Outwater EK, Bhatia M, Siegelman ES, Burke MA, [111] Nelson HA, Gilfeather M, Holman JM, Nelson EW,
Mitchell DG. Lipid in renal clear cell carcinoma: de- Yoon HC. Gadolinium-enhanced breathhold three-
tection on opposed-phase gradient-echo MR images. dimensional time-of-flight renal MR angiography in
Radiology 1997;205:103 – 7. the evaluation of potential renal donors. J Vasc Interv
[98] Mitchell DG, Crovello M, Matteucci T, Petersen RO, Radiol 1999;10(2 pt 1):175 – 81.
Miettinen MM. Benign adrenocortical masses: diag- [112] Hood MN, Ho VB, Corse WR. Three-dimensional
nosis with chemical shift MR imaging. Radiology phase-contrast magnetic resonance angiography:
1992;185:345 – 51. a useful clinical adjunct to gadolinium-enhanced
906 J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907

three-dimensional renal magnetic resonance angiog- angiography for evaluating renal artery stenosis.
raphy? Mil Med 2002;167:343 – 9. AJR Am J Roentgenol 2001;176:1213 – 9.
[113] Silverman JM, Friedman ML, Van Allan RJ. Detec- [125] Volk M, Strotzer M, Lenhart M, Seitz J, Manke C,
tion of main renal artery stenosis using phase-contrast Feuerbach S, et al. Renal time-resolved MR angi-
cine MR angiography. AJR Am J Roentgenol 1996; ography: quantitative comparison of gadobenate
166:1131 – 7. dimeglumine and gadopentetate dimeglumine with
[114] Binkert CA, Debatin JF, Schneider E, Hodler J, different doses. Radiology 2001;220:484 – 8.
Ruehm SG, Schmidt M, et al. Can MR measurement [126] Lee V, Rofsky NM, Krinsky GA, Stemerman DH,
of renal artery flow and renal volume predict the out- Weinreb JC. Single-dose breath-hold gadolinium-
come of percutaneous transluminal renal angioplasty? enhanced three-dimensional MR angiography of the
Cardiovasc Intervent Radiol 2001;24:233 – 9. renal arteries. Radiology 1999;211:69 – 78.
[115] Lee VS, Rofsky NM, Ton AT, Johnson G, Krinsky [127] Earls J, Rofsky NM, DeCorato DR, Krinsky GA,
GA, Weinreb JC. Angiotensin-converting enzyme Weinreb JC. Hepatic arterial-phase dynamic gadoli-
inhibitor-enhanced phase-contrast MR imaging to nium-enhanced MR imaging: optimization with a test
measure renal artery velocity waveforms in patients examination and a power injector. Radiology 1997;
with suspected renovascular hypertension. AJR Am J 202:268 – 73.
Roentgenol 2000;174:499 – 508. [128] Wilman AH, Riederer SJ, King BF, Debbins JP, Ross-
[116] Schoenberg SO, Knopp MV, Bock M, Kallinowski F, man PJ, Ehman RL. Fluoroscopically triggered con-
Just A, Essig M, et al. Renal artery stenosis: grading trast-enhanced three-dimensional MR angiography
of hemodynamic changes with cine phase-contrast with elliptical centric view order: application to the
MR blood flow measurements. Radiology 1997;203: renal arteries. Radiology 1997;205:137 – 46.
45 – 53. [129] Leung DA, Pelkonen P, Hany TF, Zimmermann G,
[117] Hahn U, Miller S, Nagele T, Schick F, Erdtmann B, Pfammatter T, Debatin JF. Value of image subtrac-
Duda S, et al. Renal MR angiography at 1.0 T: three- tion in 3D gadolinium-enhanced MR angiography
dimensional (3D) phase-contrast techniques versus of the renal arteries. J Magn Reson Imaging 1998;8:
gadolinium-enhanced 3D fast low-angle shot breath- 598 – 602.
hold imaging. AJR Am J Roentgenol 1999;172: [130] Lee V, Martin DJ, Krinsky GA, Rofsky NM. Gado-
1501 – 8. linium-enhanced MR angiography: artifacts and pit-
[118] Prince MR, Schoenberg SO, Ward JS, Londy FJ, falls. AJR Am J Roentgenol 2000;175:197 – 205.
Wakefield TW, Stanley JC. Hemodynamically signi- [131] Johnson PT, Halpern EJ, Kuszyk BS, Heath DG,
ficant atherosclerotic renal artery stenosis: MR angio- Wechsler RJ, Nazarian LN, et al. Renal artery steno-
graphic features. Radiology 1997;205:128 – 36. sis: CT angiography – comparison of real-time vol-
[119] Lee VS, Flyer MA, Weinreb JC, Krinsky GA, Rof- ume-rendering and maximum intensity projection
sky NM. Image subtraction in gadolinium-enhanced algorithms. Radiology 1999;211:337 – 43.
MR imaging. AJR Am J Roentgenol 1996;167: [132] Floemer F, Glombitza G, Knopp MV, Schoenberg
1427 – 32. SO, Brockmeier K, Meinzer HP. Use of virtual reality
[120] Earls JP, Rofsky NM, DeCorato DR, Krinsky GA, for MRI data of complex vascular structures. Radiol-
Weinreb JC. Breath-hold single dose Gd-enhanced oge 2000;40:246 – 55.
three dimensional MR aortography; usefulness of a [133] Sakai G, Nishida N, Yamada R, Matsuoka T. Con-
timing examination and MR power injector. Radiol- trast-enhanced MR angiography of the epigastric and
ogy 1996;201:705 – 10. hepatic regions: visibility in three-dimensional recon-
[121] Lee VS, Rofsky NM, Krinsky GA, Stemerman DH, structions. Osaka City Med J 2000;46:1 – 15.
Weinreb JC. Single-dose breath-hold gadolinium- [134] Sagami A. Evaluation of time of flight MR angiog-
enhanced three-dimensional MR angiography of the raphy for stenotic arterial lesions: including compari-
renal arteries. Radiology 1999;211:69 – 78. son of maximum intensity projection and volume
[122] Schoenberg SO, Bock M, Knopp MV, Essig M, Laub rendering technique. Nippon Igaku Hoshasen Gakkai
G, Hawighorst H, et al. Renal arteries: optimization of Zasshi 1994;54:975 – 87.
three-dimensional gadolinium-enhanced MR angiog- [135] Johnson P, Heath DG, Bliss DF, Cabral B, Fishman
raphy with bolus-timing-independent fast multiphase EK. Three-dimensional CT: real-time interactive vol-
acquisition in a single breath hold. Radiology 1999; ume rendering. AJR Am J Roentgenol 1996;167:
211:667 – 79. 581 – 3.
[123] Volk M, Strotzer M, Lenhart M, Manke C, Nitz WR, [136] Ney D, Fishman EK, Kawashima A, Robertson Jr
Seitz J, et al. Time-resolved contrast-enhanced MR DD, Scott WW. Comparison of helical and serial
angiography of renal artery stenosis: diagnostic accu- CT with regard to three-dimensional imaging of
racy and interobserver variability. AJR Am J Roent- musculoskeletal anatomy. Radiology 1992;185:
genol 2000;174:1583 – 8. 865 – 9.
[124] Masunaga H, Takehara Y, Isoda H, Igarashi T, Su- [137] Hany T, Schmidt M, Davis CP, Gohde SC, Debatin
giyama M, Isogai S, et al. Assessment of gadoli- JF. Diagnostic impact of four postprocessing tech-
nium-enhanced time-resolved three-dimensional MR niques in evaluating contrast-enhanced three-dimen-
J. Zhang et al / Radiol Clin N Am 41 (2003) 877–907 907

sional MR angiography. AJR Am J Roentgenol 1998; planar reformatting, and 3D volume-rendering post-
170:907 – 12. processing algorithms. Acad Radiol 2002;9:50 – 9.
[138] Baskaran V, Pereles FS, Nemcek Jr AA, Carr JC, [139] Cherrak I, Jaulent MC, Azizi M, Plouin PF, Degoulet
Miller FH, Ly J, et al. Gadolinium-enhanced 3D P, Chatellier G. Can the degree of renal artery stenosis
MR angiography of renal artery stenosis: a pilot be automatically quantified? Arch Mal Coeur Vaiss
comparison of maximum intensity projection, multi- 2000;93:1047 – 52.
Radiol Clin N Am 41 (2003) 909 – 929

Evaluation of renal causes of hypertension


Robert P. Hartman, MD*, Akira Kawashima, PhD, MD, Bernard F. King, Jr, MD
Department of Radiology, Mayo Clinic, 200 First Street Southwest, Rochester MN 55905, USA

Hypertension currently affects approximately pheochromocytoma, coarctation of the aorta, hyper-


60 million Americans and can lead to significant calcemia, carcinoid syndrome, central nervous system
morbidity and mortality including heart disease and tumors, and acromegaly. This article concentrates on
renal failure. Although most people suffer from essen- the renal-mediated secondary causes of hypertension.
tial hypertension, a substantial subset (5% to 10%) has
a secondary cause. Of this group of secondary causes,
renal parenchymal disease and renovascular disease Renovascular disease
are the most prevalent. Specific causes are as follows:
Renal artery stenosis
Renovascular disease
Atherosclerosis Renal artery stenosis (RAS) accounts for most
Fibromuscular dysplasia renal-mediated hypertension, likely accounting for
Renal artery aneurysm 1% to 5% of all cases of hypertension in the population
Arterial artery dissection [1]. The benefits of revascularization (either surgically
Vasculitis (polyarteritis nodosa) or angiographically) are generally accepted as a better
Takayasu’s arteritis form of long-term therapy in these patients [2].
Neurofibromatosis Although affecting a smaller group of people, the
Mid-aortic syndrome diagnosis of hypertension secondary to RAS is impor-
Arteriovenous communications tant, because it is often a correctable cause of disease.
Posttraumatic hypertension If treated, the blood pressure can be lowered or cured
Renal parenchyma disease and the kidneys and other end organs can be protected.
Nephropathy (diabetic nephropathy, glomeru- Renovascular hypertension is a renin-dependent
lonephritis, lupus nephritis, nephrosclerosis) elevation of blood pressure resulting from renal
Chronic pyelonephritis ischemia and decreased renal profusion caused by a
Tumors (juxtaglomerular cell tumor) stenotic lesion of the renal artery or its segmental
Polycystic kidney disease (autosomal dominant branches. This results in decreased perfusion of the
and recessive) glomerulus. This decreased glomerular blood flow in
Page kidney (chronic subcapsular hematoma the afferent arteriole is detected by baroreceptors in
and perirenal fibrosis) the juxtaglomerular apparatus. This results in renin
release into the bloodstream. In addition, decreased
Other nonrenal secondary causes of hypertension sodium load in the renal tubules, as a result of renal
include primary aldosteronism, Cushing’s disease, artery stenosis, is detected by the macula densa,
which in turn causes further renin release by the
juxtaglomerular apparatus. Increased renin levels re-
sult in the production of angiotensin II, which then
* Corresponding author. goes on to cause vasoconstriction of blood vessels
E-mail address: Hartman.Robert@mayo.edu and an increase in the production of aldosterone.
(R.P. Hartman). Aldosterone can then go on to produce vasoconstric-

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00071-X
910 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

tion of the blood vessels and also results in sodium Refractory hypertension (not responsive to
and water retention (Fig. 1). therapy with  three drugs)
Renal vascular hypertension from RAS has a low Renal abnormalities
prevalence within the general public. Screening of all Unexplained azotemia (suggestive of athero
patients with hypertension for RAS is not warranted. sclerotic RAS)
Careful clinical screening of patients for particular Azotemia induced by treatment with an
findings or medical histories, however, allows for the angiotensin converting enzyme inhibitor
identification of patients at higher risk of RAS [3]. Unilateral small kidney
Clinical findings associated with renal artery stenosis Unexplained hypokalemia
include the following: Other findings
Abnormal bruit, flank bruit, or both
Hypertension Severe retinopathy
Abrupt onset of hypertension before the age of Carotid, coronary, or peripheral vascular
30 years (suggestive of fibromuscular disease
dysplasia) Unexplained congestive heart failure or acute
Abrupt onset of hypertension at or after the age pulmonary edema
of 50 years (suggestive of atherosclerotic
RAS) In particular, the onset of hypertension before the
Accelerated or malignant hypertension age of 30 or after the age of 50, worsening hyperten-

Fig. 1. Pathophysiology of renovascular hypertension. (From the Mayo Foundation; with permission.)
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 911

sion despite multiple therapies, associated peripheral


vascular disease, cigarette smoking, an abdominal
bruit, or concomitant renal dysfunction in the setting
of hypertension are possible signs of atherosclerotic
RAS. The suspicion for fibromuscular dysplasia
(FMD) increases in hypertensive women under the
age of 30 or younger patients with an abdominal
bruit. Using these particular clinical findings a subset
of patients with hypertension can be selected where
the prevalence of RAS in the group can be as high as
47% [4]. These patients can then be screened appro-
priately with diagnostic imaging.
It is important to note that not all RAS infers
renal vascular hypertension. The diagnosis of renal
vascular hypertension is reserved for patients with
renin-mediated hypertension who benefit from
revascularization of the affected kidney or kidneys.
In some patients RAS may be present but they do not Fig. 2. Atherosclerotic renal artery stenosis. Digital sub-
receive any benefit from revascularization, and some traction abdominal aortogram demonstrates a high-grade
stenosis of the proximal left renal artery (arrow).
patients with essential hypertension have RAS that is
not hemodynamically significant.
For these reasons the diagnostic evaluation of development of atherosclerosis include diet, high
patients with suspected RAS can be challenging. Many cholesterol, smoking, and genetic predisposition [6].
imaging modalities have the ability to detect anatomic Renal artery stenosis secondary to atherosclerotic
stenoses of the renal arteries including catheter-di- disease may be caused by a primary lesion within the
rected digital subtraction angiography, CT angiog- renal artery or from secondary extension of plaque
raphy, color-flow Doppler ultrasonography, and MR from the abdominal aorta. Primary lesions are caused
angiography. Angiotensin converting enzyme (ACE) by eccentrically located atheromatous plaques that
inhibition renal scintigraphy images the functional usually affect the proximal third of the renal artery
changes associated with RAS rather than the anatomic (Fig. 2). Ostial lesions caused by exuberant plaque
lesion. MR angiography and Doppler sonography can within the aorta are difficult to differentiate from
also measure functional changes. Serum and renal vein renal artery plaques but have important implications
renin measurements have also been advocated in the for therapy.
evaluation of renovascular hypertension. The plaques reside beneath the intima and are a
Renal artery stenosis is the leading cause of renal mixture of cholesterol or cholesterol esters and fi-
vascular hypertension. The two most prevalent causes brous tissue with or without calcification. The fibrous
of RAS in the population are atherosclerotic disease tissue is comprised of smooth muscle cells, collagen,
and FMD. Other diseases, such as neurofibromatosis, elastin, and proteoglycans. The plaque can enlarge or
vasculitis, renal artery dissection, aortic aneurysm, or coalesce leading to luminal narrowing. This leads to
mid-aortic syndrome, account for a small percentage activation of the renin-angiotensin II pathway and
of RAS. higher systemic blood pressures.
Atherosclerosis is a progressive disease and can
lead to renal insufficiency or failure. Occlusion of the
Atherosclerotic disease renal artery also has been reported in up to 14% of
cases [7].
Atherosclerotic disease is a progressive degenera-
tive disease that affects older patients. It is the most Fibromuscular dysplasia
common disease causing RAS, accounting for ap-
proximately 70% to 80% of cases [5]. The disease Fibromuscular dysplasia is the second most com-
usually affects the entire circulatory system and can mon cause of RAS, accounting for approximately
cause other symptoms, such as angina, transient 15% to 20% of all cases. There are four major types
ischemic attacks, claudication, or mesenteric ische- of FMD. In adults the most common form is medial
mia. The lesions usually involve the origin or proxi- fibroplasia. The other forms are perimedial fibro-
mal portion of the renal artery. Risk factors for the plasia, intimal fibroplasia, and medial hyperplasia
912 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

[8]. Adventitial or periarterial fibroplasia can also of narrowing that can be accompanied by regions of
result in RAS. poststenotic dilatation with a resulting classical ap-
Medial fibroplasia accounts for about 70% of all pearance of the ‘‘string of beads’’ (Fig. 3) [8].
cases of FMD. It occurs most often in women between Perimedial fibroplasia occurs in women between
the ages of 25 and 50 years and is rare in children. It the ages of 15 and 30 years. It comprises about 15%
tends to affect the distal two thirds of the main renal to 20% of all cases. A tight stenosis of the renal artery
artery and its major branches. Other vessels in the occurs secondary to deposition of collagen in the
body, such as the carotid and vertebral arteries and the outer border of the media.
mesenteric and iliac arteries, also can be involved. Intimal fibroplasia is a rare condition that affects
Histologically the internal elastic membrane is lost children and young adults (often male), accounting for
or thinned and collagen bands replace the muscle. 2% to 5% of all cases. It results in a focal narrowing of
These bands within the renal artery lead to focal areas the renal artery or segmental arteries because of

Fig. 3. (A) Digital subtraction, catheter-directed angiogram of the abdominal aorta and branches demonstrates alternating areas
of stenosis with a beaded appearance in the main right renal artery (arrow). The changes are characteristic to fibromuscular
dysplasia. Maximum intensity projection (MIP) contrast-enhanced MR angiogram (B), three-dimensional volume rendering (VR)
CT angiogram (C), and catheter-directed selective digital subtraction angiogram (D) of the same patient.
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 913

circumferential collagen deposition in the internal the renin-angiotensin pathway leads to increased
elastic lamina. Medial hyperplasia is also rare, result- levels of circulating angiotensin II. Angiotensin II
ing from diffuse proliferation of smooth muscle. affects the kidneys by constricting the efferent arte-
Adventitial or periarterial fibroplasia causes 1% of riole resulting in higher pressures within the glo-
cases of FMD. Focal or tubular stenoses result from meruli. When an ACE inhibitor is introduced the
dense fibrous tissue surrounding the renal artery. conversion of angiotensin I to angiotensin II is
limited, lowering circulating levels and diminishing
Imaging of RAS secondary to atherosclerotic disease the result on the efferent arteriole (Fig. 4). This in
and FMD turn decreases the perfusion pressure within the
glomerulus with resultant drop in the glomerular
The imaging of RAS caused by atherosclerosis or filtration rate [11]. The filtration of the circulating
FMD is similar. The following sections are descrip- isotope is decreased (Tc 99m diethylenetriamine
tions of the many imaging modalities that are avail- pentaacetic acid) and urine flow drops leading to
able to the radiologist when a patient is referred with delayed transit time of tubular secretion agents
the possibility of RAS from either of these etiologies. (I 131 o-iodohippurate sodium, Tc 99m mercapto-
acetyltriglycine) (Fig. 5).
Functional versus anatomic imaging Different institutions use varying protocols for
The choice of modality used as the initial tool for ACE-inhibition scintigraphy. In all cases, patients
the evaluation of RAS varies within institutions based have to discontinue ACE-inhibitor medication being
on availability and physician expertise. In addition, the used to treat their hypertension. Although it is ideal to
clinical suspicion for RAS plays a role. In a patient withhold all ACE-inhibition medication for 48 hours,
with a very low suspicion of RAS, imaging of the renal most institutions withhold ACE inhibitors for only
vasculature should not be pursued. In contrast, given a 24 hours. Some institutions obtain a baseline scan
high level of suspicion for RAS, catheter-directed followed by a scan with ACE inhibition either later
contrast angiography is a legitimate choice because that day or on a following day. The 1-day protocol is as
therapeutic measures can be provided at the time of the follows. A baseline blood pressure is obtained and the
study. The group of patients with an intermediate level patient is hydrated to ensure adequate urine flow
of suspicion for RAS is those that are often referred for during the examination. The initial scan is performed
noninvasive screening examinations. and uptake and excretion curves are obtained. The
Some modalities offer direct identification of a initial dose is allowed to clear and after a number of
lesion resulting in RAS, whereas others identify hours a second blood pressure is obtained and
functional changes in the kidney. The modalities recorded. An ACE inhibitor is administered and serial
capable of detecting anatomic lesions include digital blood pressures are obtained every 15 minutes for 60 to
subtraction angiography, color-flow Doppler ultra- 90 minutes while the patient receives intravenous
sonography, CT angiography, and MR angiography. fluids. A second dose of radiopharmaceutical is then
The only modality in widespread use for the detection administered and the patient is scanned again, obtain-
of changes in renal function secondary to RAS is ing the appropriate uptake and excretion curves
ACE-inhibition renal scintigraphy, although ACE- as before.
inhibition dynamic gadolinium MR imaging is under An alternative method for the examination is a
investigation [9,10]. single scan after the administration of an ACE inhibi-
tor; however, this demonstrates a lower sensitivity and
ACE-inhibition renal scintigraphy specificity than the combined pre – and post – ACE-
Angiotensin converting enzyme inhibition renal inhibition studies. This relies on the delayed elimina-
scintigraphy images the kidneys with tubular agents, tion of isotope from the kidney to diagnose RAS,
such as Tc 99m mercaptoacetyltriglycine and I 131 rather than the change in the elimination of isotope
o-iodohippurate sodium, or glomerulofiltration agents, from baseline to post – ACE-inhibitor scans. If the scan
such as Tc 99m diethylenetriamine pentaacetic acid is normal, only one dose of radiopharmaceutical needs
(DTPA). Currently, Tc 99m mercaptoacetyltriglycine to be administered and this cuts down on examination
(MAG3) is the most commonly used agent. The scan times. If equivocal findings are present on the single
is performed as an outpatient examination after the scan, a baseline examination can then be performed
patient’s use of ACE inhibitors (ie, captopril) has after an appropriate interval to allow for the ACE-
been discontinued. inhibition to wear off.
In a patient with unilateral RAS, the affected The ACE-inhibition scintigraphy has a reported
kidney increases the output of renin, which through sensitivity of 80% and specificity of near 100% for
914 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

Fig. 4. Physiologic effects of renal artery stenosis and angiotensin converting enzyme inhibitor (ie, captopril) on glomerular
filtration. (From the Mayo Foundation; with permission.)

the detection of RAS [12]. ACE-inhibition scintig- A positive scan is sufficient evidence to proceed
raphy provides a functional rather than anatomic to angiography and intervention if a lesion is iden-
diagnosis and is recommended when an intermediate tified. Predicted improvement in blood pressure after
clinical suspicion exists or there is a desire to deter- intervention in these patients has a reported accuracy
mine whether a known RAS is functionally signifi- of 90% to 98% [13].
cant. In addition, there is good correlation between
the findings on renal scintigraphy and results of Doppler ultrasonography
treatment. Pretherapy scans act as good baseline Since the introduction of duplex Doppler color-
examinations, and residual or restenosis of the artery flow ultrasound the usefulness of ultrasound for the
can be detected after intervention has occurred. detection of RAS has improved. Ultrasound is a less
The ability to detect disease with renal scintig- expensive noninvasive test that can be performed on
raphy is limited in patients with renal insufficiency, patients without necessitating the discontinuation of
which is prevalent within older patient populations their antihypertensive medications. The usefulness of
with atherosclerosis, and in patients with bilateral ultrasound remains limited, however, secondary to
disease. Most often in bilateral disease there is an operator-dependent issues; technical failure (body
asymmetry of disease that can be detected; however, habitus, bowel gas); and inability to image small
occasionally bilateral symmetric disease can be accessory arteries. Despite these limitations, ultra-
missed. For these reasons it is most beneficial to sound has become a leading modality in screening
acquire pre – and post – ACE-inhibition studies. for RAS.
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 915

Fig. 5. Unilateral renal artery stenosis. (A) Tc 99m MAG3 angiotensin converting enzyme inhibition (captopril) renogram images
from 1 minute through 20 minutes, including postvoid image, demonstrates delayed progression of tracer activity in the left
kidney with delayed cortical uptake (short arrow) and excretion (long arrow), characteristic of left renal artery stenosis.
(B) Cortical perfusion curve demonstrates the quantitative measure of the delayed tracer activity in the left kidney relative to the
aorta and the right kidney. (C) Excretion curve demonstrates the quantitative measure of the prolonged tracer activity in the left
kidney relative to the right kidney.
916 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

The detection of RAS with ultrasound requires a of these examinations. In addition to nonvisualization
complete examination of the kidneys including gray- of the main renal arteries, 14% to 24% of patients
scale images of the kidney size and contour and color have accessory renal arteries that often are undetected
Doppler and spectral tracings of the vessels. The by ultrasound [18]. In the authors’ practice, this
examination of the renal vasculature includes images technique has a sensitivity and specificity of 85%;
of the main renal artery and intrarenal segmental however, the limitations of technical failures and
arteries. Detection of RAS when evaluating the main nonvisualized accessory arteries are significant.
renal arteries depends on an increased peak systolic An additional method of sonographic evaluation
velocity of blood flow through the stenotic segment for RAS focuses on changes in the segmental renal
and changes seen in the caliber of the color flow arteries downstream from the stenosis. Although the
through the artery and Doppler aliasing. As the visualization of the main renal arteries is variable, the
stenosis progresses, the cross-sectional diameter of segmental arteries within the kidneys are usually easy
the lumen decreases. This in turn leads to an increase to routinely identify and interrogate. These arteries
in the velocity of blood traversing the segment. are too distant from the stenosis to exhibit increased
The upper limit of peak systolic velocity that indi- velocities but rather have dampened velocity wave-
cates a significant stenosis varies in the literature. forms. Specifically, the segmental arteries can exhibit
Currently, a peak systolic velocity greater than 180 to a tardus (late)-parvus (small) waveform in patients
200 cm/second is commonly accepted as this limit with a more proximal RAS.
(Fig. 6A) [14,15]. Additionally, a relative velocity The normal velocity waveform within a segmental
ratio comparing the aorta with the renal artery has artery has a rapid systolic upstroke with an early
been used. In this instance a renoaortic peak systolic systolic peak. The velocity waveform of all renal
velocity ratio of greater than 3.5 is indicative of a arterial vessels is a low-resistance waveform with
hemodynamically significant stenosis [16]. In addi- forward flow in diastole. Quantitatively, the segmen-
tion to velocity change, a focal stenosis often causes tal arteries can be evaluated with several different
turbulent flow within the artery that can be identified parameters: the acceleration time, acceleration index,
as aliasing on the color Doppler images. and the resistive index. The acceleration time is the
To use these criteria it is necessary to image the time from the start of systole to peak systole, and is
entire length of the main renal arteries. This unfortu- normally less than 70 milliseconds. The acceleration
nately is not as easy as it might seem. Technical index is the slope of the systolic upstroke and is
failure to image the renal artery along its length normally at least 3.5 m/second [19]. The resistive
commonly occurs in obese patients and patients with index is a ratio of the systolic and diastolic velocities
a large amount of bowel gas. The technical failure within the segmental arteries. It is calculated with the
rates range from 4% to 42% in the literature [17]. The use of the following equation: [1-(end-diastolic ve-
examination is operator-dependent, and failure rates locity/ maximal systolic velocity)]. A normal value
are lower at institutions that perform larger numbers for the resistive index is usually less than 0.7 [17].

Fig. 6. Renal artery stenosis. (A) Duplex ultrasonogram of the main right renal artery demonstrates elevated peak systolic
velocities (5.53 m/second, normal < 1.8 m/second) consistent with a high-grade stenosis. (B) Duplex ultrasonogram of the
segmental renal artery in the upper pole of the right kidney in the same patient demonstrates a tardus-parvus waveform distal to
the main renal artery stenosis. Note the acceleration time (dT) is prolonged (179 milliseconds, normal < 70 milliseconds) and the
acceleration index is lower (2.36 m/second, normal > 3.5 m/second).
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 917

In contrast, the segmental arteries downstream correlation with significant stenoses [21]. These
from a hemodynamically significant RAS have a examinations, however, took many minutes to per-
delayed systolic upstroke with decreased overall flow. form with image quality suffering from respiratory
This is the aforementioned tardus-parvus waveform. motion. Overgrading of stenoses caused by the
Acceleration time is longer while the acceleration dephasing artifact also occurred. In addition, only
index is lower than normal secondary to the delayed the proximal renal arteries could be imaged, which
systolic peak. The resistive index is greater because often excluded identification of stenoses caused by
of lower amplitude of the systolic peak (Fig. 6B). FMD, which tend to be more peripheral.
This method by itself also has been shown to have In the past few years gadolinium-enhanced three-
sensitivity and specificity greater than 80% for the dimensional spoiled gradient echo imaging during a
detection of RAS, whereas not having as high a single breathhold has become more widely used. The
degree of technical failure [19]. images do not suffer from respiratory motion degra-
Recent studies using a combined approach of dation, and the three-dimensional image data set
imaging both the main renal arteries and the segmental allows for viewing of the renal arteries in an infinite
renal arteries have reported a technical failure rate of number of projections. Gadolinium bolus timing and
0% with 96% sensitivity and 98% specificity com- the advent of elliptical centric k-space sampling have
pared with angiography [17]. This combined approach allowed for the imaging of the renal arteries at peak
is likely the best way of evaluating for RAS with arterial enhancement during the first pass while
Doppler ultrasound presently. In the future ultrasound limiting venous contamination of the image and
examinations following ACE inhibition to enhance the further reducing the effects of respiratory motion
tardus-parvus waveform, use of echocontrast media, (Fig. 7) [22].
and future generations of sonographic machines may Finally, smaller fields of view are being used to
increase the usefulness of ultrasonography as a screen- increase the spatial resolution of the images allowing
ing modality beyond where it is now. for better grading of the degree of stenosis and aiding
Besides imaging the anatomic stenosis attempts in the detection of peripheral stenoses and the visual-
have been made using sonographic criteria to deter- ization of accessory renal arteries. This has been
mine which patients with RAS benefit from revascu- shown to have a sensitivity of 97% and specificity
larization. A recent study suggests that renal function of 92% compared with intra-arterial angiography for
and blood pressure did not improve in patients with a the detection of RAS including distal main and
resistive index greater than 0.80 in an effected kidney segmental arteries [23]. Within the study group all
despite revascularization [20]. This is possibly be-
cause of changes within the renal parenchyma and
vessels, such as glomerulosclerosis or nephrosclero-
sis, from long-standing hypertension. Further studies
regarding this are necessary.

MR angiography
MR angiography has made significant progress in
its use as a screening examination for RAS in the past
few years in part because of improvements in gradient
systems, the advent of breathhold imaging sequences,
and centric k-space phase encoding. The examination
continues to have limitations because of lack of
widespread availability of capable MR imaging scan-
ners; costs; and patient issues, such as claustrophobia.
In the past phase contrast and time-of-flight MR
angiography of the renal arteries was used for the
detection of RAS. The diagnosis was based on
imaging the anatomic narrowing in the vessel (time-
of-flight and phase contrast) and signal loss caused by
dephasing of the blood from turbulent flow in the Fig. 7. Atherosclerotic renal artery stenosis. Gadolinium-en-
poststenotic renal artery (phase contrast). Dephasing hanced, MR angiogram of the abdominal aorta and renal
of blood within a given artery on phase-contrast MR arteries demonstrates bilateral, proximal renal artery stenoses
angiography has been shown in the past to have a (arrows) caused by atherosclerotic renal artery disease.
918 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

segmental artery stenoses and middle to distal main increased the spatial resolution of these studies,
artery stenosis caused by FMD were depicted [23]. which was already greater than the spatial resolution
MR angiography is continuing to improve in the of MR angiography [24]. Image sets are recon-
detection of RAS. In addition to anatomic depiction structed to submillimeter slice thickness during
of stenoses the possibility of using dynamic gado- reprocessing. The image data then can be viewed
linium images of the kidneys to detect functional on a capable workstation in an infinite number of
changes similar to those evaluated by ACE-inhibited projections given the three-dimensional acquisition.
scintigraphy may be possible [9,10]. Ultimately, this Different three-dimensional rendering techniques, in-
may provide similar predictive value regarding the cluding maximum intensity projection and volume
likelihood of improvement in renal function and rendering, have been studied in recent years for their
blood pressure in patients with diagnosed RAS. This individual ability to detect RAS [25].
would allow for a better selection of patients to Maximum intensity projection algorithms produce
undergo revascularization of the kidney. images by establishing the maximum voxel intensity
along a ray within the data set. This allows for
CT angiography differentiation between intra-arterial contrast and ec-
With the advent of slip ring, single-detector heli- centric calcified plaque along the arterial wall, and
cal CT scanners it became possible to perform 40 different attenuations within the kidneys themselves.
contiguous 1-second tube rotations within a single Some of the three-dimensional relationships, how-
breathhold. This combined with continuous table ever, can be lost. When viewing maximum intensity
motion during the examination allowed for the projection images it is important to view the original
acquisition of volumetric imaging data from the data set in multiple projections to ensure that plaque
abdominal aorta and its major branches. Recent is seen adjacent to, rather than overlying, the lumen.
availability of multidetector helical CT scanners has This helps decrease the overestimation of stenosis

Fig. 8. Noncalcified atherosclerotic renal artery stenosis. Three-dimensional maximum intensity projection (A) and volume
rendering (B) CT angiograms demonstrate a focal noncalcified stenosis in the main left renal artery (arrows). Note the
poststenotic dilatation of the renal artery.
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 919

severity. The sensitivity of maximum intensity pro- The greatest advantage of catheter-directed angi-
jection images is approximately 94% with a speci- ography over the previously discussed modalities is
ficity of 87% (Fig. 8) [25]. the ability to intervene and correct the stenosis if
Volume rendering is an interactive three-dimen- identified. The treatment options for revascularization
sional-rendering algorithm that computes a volumetric are discussed later. Disadvantages include the use of
image that contains all attenuation values. To view the iodinated contrast media and its inherent risks of
renal vessels, however, the display must be manipu- allergic reaction and relative contraindication in
lated to remove obscuring structures. To do this patients with renal insufficiency. In addition, the
subjective optimization of the display is required procedure is invasive and caries a reported mortality
including window width, level, and brightness. As in rate of 0% to 2%. Other complications, although rare,
maximum intensity projection images the image data include contrast-induced renal failure, atheroemboli,
can be viewed in multiple projections and calcified pseudoaneurysm, and hematoma. Currently, only
plaque in the artery is easily distinguished from patients with a high suspicion of RAS should proceed
luminal contrast. Volume rendering is better than to intra-arterial angiography as the first diagnostic
maximum intensity projection images in maintaining tool given the potential complications.
the three-dimensional vascular relationships. The sen-
sitivity of volume rendering for the detection of RAS is Treatment of RAS
similar to maximum intensity projection, whereas the
specificity is better, approximately 99% (Fig. 9) [25]. The treatment options in patients with RAS
Accessory arteries to the kidneys can arise any- caused by atherosclerosis or FMD are generally
where along the aortoiliac course from T11 to L4. percutaneous intervention versus surgery for revas-
Ninety percent of these, however, occur within 17 mm cularization of the kidney. The patient’s health status,
cephalad to the main renal artery and 70 mm caudal. and etiology and severity of the RAS, including
For this reason, images should be prescribed to vascular disease in the abdominal aorta and mesen-
include as much of the abdominal aorta as possible. teric arteries, must be considered in determining the
CT angiography is a good test for detecting accessory proper intervention. In general, percutaneous trans-
arteries when they are present. luminal renal artery angioplasty (PTRA), with or
The limitations of CT angiography include the use without stenting, has become a commonly accepted
of iodinated contrast material and ionizing radiation. initial therapy.
A substantial number of patients with suspected RAS
have renal insufficiency, a relative contraindication to Percutaneous transluminal renal artery angioplasty
the use of iodinated contrast media. In addition, there Since its introduction in 1978, PTRA has gained
is the possibility of allergic reactions when adminis- steady acceptance and has produced increasingly
tering iodinated contrast media. For these reasons, CT positive results. This is because of advancements in
angiography may not be the best first choice as a soft and hydrophilic-coated wires, and catheter and
screening test for RAS. balloon designs. The procedure consists of identify-
ing the stenotic segment in the renal artery with an
Intra-arterial angiography injection of contrast material, traversing the segment
Catheter-directed intra-arterial angiography re- with a soft guidewire, positioning an expandable
mains the gold standard for the detection of RAS. It balloon across the segment, and inflating the balloon
is most commonly performed using a Seldinger tech- to expand the diameter of the segment. In atheroscle-
nique to gain access to a femoral artery. Using a small rotic disease the mechanism of therapy consists of
catheter (4 to 5F catheter) iodinated contrast, CO2, or fracturing of the atheroma, tearing of the intima and
gadolinium chelate is injected into the abdominal aorta media, and dilatation of the adventitia. In contrast,
at the level of the renal arteries. Multiple projections PTRA of FMD results in fracturing of the bands or
should be obtained to ensure complete visualization of webs that cause the focal luminal stenosis.
the entire course of the main renal arteries. This helps Percutaneous transluminal renal artery angioplasty
limit the chance of overlooking a short ostial stenosis. is now the accepted therapy for patients with FMD.
Grading of the stenosis is done by comparing the This is caused in part by the fact that FMD is not a
luminal diameter at the stenosis with the diameter of systemic disease and when successfully treated the
the normal-caliber renal artery adjacent to the stenosis. lesions do not tend to recur. The technical success
Digital subtraction angiography also can be performed rate for PTRA in treating FMD is about 90% with a
in a similar manner. This can allow for the use of therapeutic benefit in 70% to 90% of patients [26,27].
smaller amounts of contrast material. Patients with FMD tend to be younger and have
920 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

Fig. 9. Atherosclerotic renal artery stenosis. Maximum intensity projection (A) and volume rendering (B) three-dimensional
CT angiograms demonstrate a calcified atherosclerotic plaque in the main right renal artery (short arrow) and a noncalcified
stenosis in the main left renal artery (long arrow). (C) Axial reformat image showing the eccentric plaque in the proximal right
renal artery. (D) Digital subtraction abdominal angiogram demonstrates the focal stenosis in the left main renal artery
corresponding to the CT angiographic findings (arrow). The right renal artery appears normal.
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 921

fewer comorbid conditions than those with athero- Surgery


sclerotic disease, and this helps limit the number of Surgical correction of RAS includes aortorenal or
complications. The complication rate ranges from 2% alternative arterial bypass procedures, unilateral ne-
to 10% and includes dissection, thrombosis, and rup- phrectomy, endarterectomy, and atherectomy. Bypass
ture of the renal artery. procedures are the most common type of surgical
Atherosclerotic vascular disease accounts for most treatment. The technical success rate for surgical
RAS, likely 70% to 80% of all cases [5]. Atheroma- intervention is high with a restenosis rate usually less
tous plaques filling a portion of the native renal artery than 10% [31]. In patients with atherosclerotic vas-
lumen cause the stenosis. It is a degenerative disease cular disease affecting the abdominal aorta and caus-
and effects older patients. In addition, it is not limited ing RAS, a surgical procedure including simultaneous
to the renal circulation and tends to progress and replacement of the aorta and revascularization of the
recur. Because of these factors the technical success kidney may be the best alternative. The rates of cure
rate for PTRA is lower in this group of patients or improvement in blood pressure following surgical
relative to those with FMD. The technical success revascularization are excellent. Clinical benefit has
rate generally reported is 80% [28]. been reported in 60% to 90% [31,32]. In addition, the
The success rate of PTRA can be evaluated in a benefit to renal function is observed in more than
number of ways. The most commonly studied is 80% with improved function in 35% to 65% [7].
clinical benefit following PTRA. Approximately 70% Surgery does carry an increased risk of morbidity
to 80% of patients with unilateral atherosclerotic renal and mortality relative to percutaneous procedures. As
artery stenosis can expect beneficial results in blood previously mentioned, the complication rate can be
pressure control following PTRA [2]. Other modes of 31%. Mortality rates range from 3% to 20% [5].
evaluation include primary and secondary patency
rates, but these have not been extensively studied.
Takayasu’s arteritis
The complication rate of PTRA in atherosclerotic
disease is higher than in FMD. These range from 10%
Takayasu’s arteritis is a rare disease that affects
to 13% and include renal artery thrombosis, dissec-
the aorta and main branches. It is divided into a
tion or rupture, embolization, myocardial and cere-
number of types according to the region of the aorta
bral infarction, renal insufficiency, pseudoaneurysm,
and branch vessels affected. When the renal arteries
and hematomas. The mortality of the procedure is up
are affected, hypertension can be a result because the
to 4% [2].
disease process causes arterial wall thickening and
A subset of RAS caused by atherosclerotic vas-
resultant narrowing of the arterial lumen. The histo-
cular disease is ostial lesions. Previously these were
logic changes in the arterial wall are caused by
believed to be refractory to PTRA. It was the opinion
transmural disorganization and fibroplasia.
of many that dilating the aortic wall in addition to the
CT and MR imaging of the aorta and renal arteries
plaque is more difficult and the risk of dislodging
including angiographic sequences can demonstrate
plaque within the aorta is greater. Despite this a 58%
the wall thickening in addition to the resultant lumi-
clinical benefit was recently reported following
nal narrowing (Fig. 10). Intra-arterial angiography
PTRA (without stents) of ostial lesions [29].
continues to be used in cases of suspected Takayasu’s
The use of expandable metallic stents in treating
arteritis, because therapy can be attempted at the time
RAS is currently under investigation. The stents may
of the diagnostic procedure.
help in the treatment of complicated or recurrent
Treatment of Takayasu’s arteritis with percuta-
stenoses, including ostial lesions. Recent reports have
neous dilatation has met with mixed results. Initial
shown a 96% technical success rate and a 64%
technical success rates can be as high as 95% but
clinical benefit in patients with ostial lesions treated
patients often ultimately require surgical interven-
with angioplasty and a stent [30].
tion [33].
The success of PTRA versus surgical intervention
has only had limited study. In one report the primary
patency, secondary patency, and clinical benefit of Neurofibromatosis
patients with unilateral atherosclerotic RAS were
similar between PTRA and surgery. The complica- Neurofibromatosis is a group of diseases that are
tions following PTRA (17%), however, were con- hereditary disorders of ectodermal origin. A number
siderably lower than following surgery (31%) [2]. of types of neurofibromatosis have been described.
PTRA continues to be a safe and effective procedure Von Recklinghausen’s disease, or neurofibromatosis
for the treatment of RAS. type 1, is an autosomal-dominant disease that can
922 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

Fig. 10. Renal artery stenosis in Takayasu’s arteritis. (A, B) Axial CT images at the level of the renal arteries performed with
contrast demonstrate circumferential wall thickening involving the abdominal aorta (long arrow) and the proximal renal arteries.
Note the high-grade luminal narrowing of the left renal artery near their origins (short arrow).

affect the renal arteries. Hypertension in neurofibro- arterial disease can preclude permanent alleviation of
matosis is often caused by an associated pheochro- the hypertension and medical therapy may be neces-
mocytoma. In patients under the age of 18, however, sary for long-term blood pressure control.
renal vascular-mediated hypertension is seven times
more likely than pheochromocytoma [34 – 36]. Midaortic syndrome
Vascular neurofibromatosis can result in hyperten-
sion caused by an aortic coarctation, extrinsic com- Midaortic syndrome is an entity caused by a
pression of the renal arteries, or lesions intrinsic to the nonspecific arteritis affecting the abdominal aorta
renal arteries. Neurofibromatosis has distinct effects and occasionally the major branches. It is often
on both the large vessels and smaller branches. Large present at birth and may progress causing sympto-
vessels are commonly surrounded by neurofibroma- matic changes in young adults. The disease can occur
tous or ganglioneuromatous tissue. In addition, either from hypoplasia of the abdominal aorta with
disorganized growth of the media and intimal pro- tubular stenosis of the renal arteries or from true
liferation occurs. In the smaller vessels a vasculitis coarctation of the aorta [38].
consisting of intimal thickening, disorganized smooth The histologic lesion may be severe intimal fibro-
muscle growth, and elastic tissue growth may be plasia or a transmural aortitis involving the adventitia
present. These changes can result in areas of stenosis, and resulting in thickening of the entire wall of the
aneurysms, and occasionally dissection of the renal aorta. Consequently, CT or MR imaging can be useful
arteries. True external compression from local neuro- modalities in the detection of the disease. As in
fibromas is rare. In neurofibromatosis, stenosis of the Takayasu’s arteritis the images can detect the wall
main renal artery is often associated with stenosis of thickening, and the angiographic portion of the
the intrarenal branches. examination can depict the luminal narrowing. It
CT or MR imaging of the abdomen including may be difficult to distinguish the imaging findings
angiographic series is likely the best option for of midaortic syndrome from Takayasu’s aortitis
radiologic evaluation. This allows for the detection and neurofibromatosis.
of aortic coarctation or RAS, whether it is caused by The treatment of midaortic syndrome is usually
extrinsic compression or intra-arterial disease. surgical, because percutaneous results have been
Treatment options for patients with neurofibro- poor [38].
matosis 1 and renal vascular hypertension include
PTRA, which has been shown to have good initial Renal artery aneurysm and dissection
results and benefits. Previous studies, however, have
suggested a restenosis rate ranging between 15% and Renal artery aneurysms can occur from a num-
60% [37]. Surgical intervention with renal artery ber of causes. They can be congenital, mycotic,
bypass is also an option. In both cases, the intrarenal traumatic, atherosclerotic, or vasculitic. The most
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 923

Fig. 11. Renal artery aneurysms. (A) Superior to inferior view of three-dimensional axial maximum intensity projection image
through the renal hila from a CT angiogram demonstrates bilateral renal artery aneurysms in the renal hila (arrows). (B) Curved
planar reformatted image from the same patient.

common cause is traumatic, either from blunt or extension into the renal artery surgical repair of the
penetrating trauma. In some cases aneurysms have aorta and renal revascularization are necessary. In
been associated with hypertension (Fig. 11). It is isolated renal artery dissection the use of PTRA and
debatable whether the aneurysm is the cause or stents may be sufficient.
the result of the hypertension. If the aneurysm is the
cause it may be on the basis of altered flow within the
renal artery, external compression of the artery, or Polyarteritis nodosa
from renal embolization. Surgical intervention should
be reserved for patients in whom there is a strong Polyarteritis nodosa is an arteritis of autoimmune
belief that the aneurysm and hypertension are related pathogenesis that tends to effect medium-sized and
or in patients where there is a concern of aneurysm small arteries in the body. Arteries anywhere in the
rupture [6]. body can be affected, but the kidneys are the most
Renal artery dissection can occur either from commonly affected sites. The disease is usually
extension of an aortic dissection or within the renal bilateral but asymmetric resulting in transmural fi-
artery alone. Isolated renal artery dissection may be brinoid necrosis and surrounding inflammation about
associated with vasculitis; neurofibromatosis; Ehlers- the vessels. The vascular abnormalities can lead to
Danlos syndrome (type IV); and blunt or iatrogenic focal areas of ischemia or infarction in the kidneys.
trauma. The dissection leads to a narrowing of These foci are likely a source of renin hypersecretion
the lumen and predisposes the artery to thrombosis. leading to hypertension. Although most patients with
This results in RAS or occlusion and can lead to polyarteritis nodosa are eventually hypertensive,
hypertension secondary to the renin-angiotensin – me- hypertension is rarely an initial finding. Treatment
diated pathway. consists of corticosteroids and other immunosup-
CT angiography of the aorta and renal arteries pressants resulting in a 5-year survival rate of
is commonly used in the evaluation of aortic dis- approximately 80% [39].
section. The study not only allows for the detection The typical angiographic findings of polyarteritis
of the dissection, it also can be used to evaluate the nodosa include abrupt angulations in the smaller
main abdominal arteries to see if any arise from the parenchymal renal arteries with irregularly margin-
false lumen. Dissection extending into the major ated lumens and multiple tiny aneurysms present and
abdominal branches can be detected. In addition, scattered within the kidneys (Fig. 12). The disease is
the scan provides anatomic detail that can aid in often segmental in nature; areas of diseased kidney
surgical planning. can be adjacent to normal parenchyma. CT angiog-
Treatment requires either surgical or percutaneous raphy and MR angiography cannot demonstrate these
intervention. In the case of an aortic dissection with changes in the smaller vessels and conventional
924 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

Angiography demonstrates the communication


and enlarged vessels but also allows for the visualiza-
tion of the pathognomonic early contrast filling of
the renal veins during the arterial injection. This
phenomenon can also now be imaged with CT using
multiphase renal angiogram protocols that include an
early arterial phase [40].

Posttraumatic stenosis

Posttraumatic stenosis of the renal arteries is


usually the result of blunt trauma. The mechanism
is caused by shearing injury of the artery between the
relatively fixed proximal portion and the less fixed
middle third. This results in subintimal dissection or
hematoma with narrowing or occlusion of the lumen.
In cases where the injury does not resolve, the lesion
can progress to a permanent stenosis and hyperten-
sion can develop.
Depending on the severity of the initial trauma the
evaluation of the patient can differ. Often CT is
performed in this setting and in the case of a complete
occlusion a nonfunctioning kidney can be seen. In
patients with a posttraumatic stenosis, however, the
Fig. 12. Polyarteritis nodosa. Selective injection of the right initial study can be normal in appearance. Catheter-
renal artery during an intra-arterial digital subtraction angio- directed angiography is only performed in patients with
gram demonstrates microaneurysms (arrows) of the intra- abdominal or pelvic injuries that necessitate its use.
renal arteries. CT angiography, MR angiography, or catheter-
directed angiography can be useful in the evaluation
of a patient with an acute onset of hypertension
angiography is recommended in patients suspected of following blunt traumatic injury. RAS discovered in
having polyarteritis nodosa. this instance, however, can be difficult to differentiate
from atherosclerotic disease or even FMD.
Arteriovenous communications

Most arteriovenous communications are acquired Renal parenchymal causes of hypertension


either iatrogenically or from penetrating trauma.
Rarely spontaneous arteriovenous communications Renal parenchymal causes of renal-mediated hy-
can occur within a neoplasm. Congenital arterio- pertension account for a very small percentage of
venous communications, or cirsoid aneurysms, usu- cases. Parenchymal diseases associated with hyper-
ally manifest between the ages of 20 and 30 years, tension include glomerulonephritis; nephrosclerosis;
with hypertension present in 25% of patients [6]. diabetic nephropathy and chronic pyelonephritis;
Hypertension in these patients is likely caused by tumors, such as renal cell carcinoma and juxtaglo-
a steal phenomenon where the renal parenchyma merular cell cancer; polycystic kidney disease; and
distal to the arteriovenous communication receives perirenal hematomas (Page kidney).
less blood flow. This leads to ischemia and excessive
renin secretion. Glomerulonephritis
The evaluation of suspect arteriovenous commu-
nications can be done with ultrasonography, CT, or Glomerulonephritis is a disease that affects the
catheter-directed angiography. Ultrasound can image glomeruli of the kidneys. Acute glomerulonephritis
the communication demonstrating high-velocity tur- has a number of etiologies including infectious en-
bulent blood flow within the communication. Renal tities; systemic diseases, such as lupus or Good-
arteries feeding the communication and the draining pasture’s syndrome; or primary glomerular diseases.
renal veins are enlarged. In the acute phase of the disease the damage to the
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 925

glomeruli results in proteinuria from leaky blood Diabetic nephropathy


vessels and a decrease in glomerular filtration rate.
The decreased filtration rate results in fluid and salt There is a high prevalence of hypertension in renal
retention that can lead to hypertension [6]. In most failure patients, depending on the type of nephropathy
cases an acute infectious glomerulonephritis resolves. and the severity of renal failure [41]. Renal insuffi-
Glomerulonephritis from other causes, and occa- ciency was thought to be a contributing factor in 1.8%
sionally secondary to an infectious etiology, however, of patients with hypertension [42]. Diabetes is the most
can progress to chronic glomerulonephritis. common cause of end-stage renal disease in the
In chronic glomerulonephritis the damage to the western world [43]. The damage to the kidneys is
glomeruli continues over time with a slow progres- believed to be a combination of hypertension and
sion of disease. This can eventually lead to renal nonhemodynamic effects of angiotensin II and aldo-
failure. Hypertension, caused by fluid retention and sterone on the kidney. The angiotensin II and aldoste-
occasionally renin hypersecretion, continues through- rone are implicated in the formation of
out the course of the disease. The diagnosis of tubulointerstitial fibrosis and glomerulosclerosis
glomerulonephritis is on the basis of renal biopsy. [44]. The prevalence of hypertension in patients with
diabetic nephropathy is 87%. The diagnosis of diabetic
Nephrosclerosis nephropathy is on the basis of renal biopsy.

Hypertension from any source can result in Chronic pyelonephritis


changes within the renal arterioles. Initially there is
constrictive effect on the arterioles followed by Chronic pyelonephritis is an interstitial nephritis
hypertrophy of the muscular walls. These changes caused by an infectious or inflammatory etiology.
progress over time leading to increased vascular The nephritis is characterized by an inflammatory
resistance within the kidney itself. Although this cellular infiltrate within the renal parenchyma. The
may not be a direct cause of hypertension these most common cause of chronic pyelonephritis is
changes may play a role in the lack of benefit seen severe vesicoureteral reflux and is often seen in
in some patients with corrected RAS. In patients with children. Other causes are anatomic abnormalities in
chronic hypertension caused by RAS, revasculariza- the kidneys that predispose the kidneys to repetitive
tion does not consistently produce a benefit [5]. Once infection. These include calculi, obstruction, or a
nephrosclerosis is present, the increased vascular neurogenic bladder [45].
resistance within the kidney is not corrected by The disease can be unilateral or bilateral and is
revascularization of an affected kidney. usually segmental. In chronic pyelonephritis from

Fig. 13. Chronic pyelonephritis. (A) Enhanced CT axial image obtained through the upper pole of the kidneys during the arterial
phase demonstrates focal regions of parenchymal scarring (arrow). There was no evidence of RAS on the three-dimensional
angiogram images (not shown). (B) Image obtained later in the examination during the excretory phase demonstrates underlying
caliceal clubbing and parenchymal scarring (arrow). Findings are characteristic of chronic pyelonephritis.
926 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

diologic evaluation should consist of thin-section en-


hanced CT of the kidneys in the search for a solid mass.
Treatment of juxtaglomerular cell tumors consists
of complete surgical excision. This commonly leads
to remission of the hypertension assuming that
underlying hypertensive changes within the renal
arterioles has not occurred as described in the section
on nephrosclerosis.
In addition to juxtaglomerular cell tumors, hyper-
tension has been associated with renal cell cancer on
occasion. This is also caused by the release of renin
from the tumor [46]. The percentage of renal cell
carcinoma that produces sufficient amounts of renin
to cause hypertension is not known.

Polycystic kidney disease


Fig. 14. Autosomal-dominant polycystic kidney disease.
Coronal T2-weighted fast spin echo MR image through the Autosomal-dominant polycystic disease kidney
kidneys demonstrates multiple bilateral hyperintense lesions results in the replacement of the normal renal paren-
in both kidneys consistent with renal cysts. The patient was chyma with multiple cysts. As implied by its name the
in their 20s and had a clinical diagnosis of autosomal- disease is hereditary, but has a wide range of pheno-
dominant polycystic kidney disease with mild hypertension. typic presentations. In some patients much of the renal
parenchyma becomes replaced and can lead to end-
stage renal disease. Hypertension has been shown to
occur more commonly in patients with autosomal-
vesicoureteral reflux, the polar regions of the kidney dominant polycystic disease kidney than their age-
are most often involved. The changes in the kidney matched controls. Hypertension in autosomal-domi-
are caused by parenchymal scarring, which results in nant polycystic disease kidney patients usually devel-
focal areas of parenchymal loss and blunting of the ops by the third or fourth decade, but can appear in
subjacent calyx (Fig. 13). The disease often is childhood or adolescence. The underlying cause of
asymptomatic for a number of years.
CT, ultrasound, and MR imaging are capable of
depicting the changes in the renal contour associated
with this disease. In addition, voiding cystoure-
thrograms are helpful in the pediatric population to
document vesicoureteral reflux. In cases of chronic
pyelonephritis caused by reflux, reimplantation of
the ureter can resolve the reflux and stop further
renal damage.

Tumors

Juxtaglomerular cell tumor, or reninoma, is a rare


tumor of the kidneys that produces renin and leads to
hypertension. It is believed to be benign in nature and
often is only a few centimeters in diameter. The
tumor occurs in patients under the age of 20 up to
Fig. 15. Chronic subcapsular hematoma (Page kidney).
50% of the time.
Enhanced CT scan through the midportion of the right kidney
Serum renin levels are elevated leading to second-
demonstrates a subcapsular thick-walled low-attenuation
ary hyperaldosteronism and hypokalemia in addition fluid collection with peripheral calcification in the wall
to hypertension. Other tumors, such as Wilms’ tumors (arrow). The fluid collection displaces the kidney anteriorly
and renal cell carcinoma, and a variety of nonrenal and compresses the renal parenchyma. The patient had a prior
diseases can be associated with increased serum renin history of blunt abdominal trauma. The patient’s hypertension
levels, so this measurement alone is not specific. Ra- improved after surgical removal of organized hematoma.
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 927

the hypertension is unknown but may be multifacto- juxtaglomerular cell tumor, but a significant number
rial including increased sodium retention, vascular of patients with essential hypertension also exhibit
compression by cysts, and increased renin-angiotensin elevated renin levels. In addition, other diseases may
II levels. The most likely cause is that increased renin result in elevation of serum renin levels.
excretion from the kidneys results from increased
vascular resistance and focal parenchymal ische-
mia secondary to compression from the renal cysts Summary
(Fig. 14) [47].
There are many renal causes of hypertension.
Subcapsular hematoma Although RAS is the most common, other renal
lesions can result in hypertension. Any evaluation
Subcapsular hematomas are located between the of the kidney for hypertension should take all of these
renal parenchyma and the fibrous renal capsule. potential renal etiologies into consideration.
These can occur from many causes including trauma
or following a renal biopsy. In addition, renal tumors,
such as renal cell carcinoma or angiomyolipomas,
can spontaneously hemorrhage [40]. If there is a References
sufficient volume of blood in the subcapsular space
[1] Vasbinder GB, Nelemans PJ, Kessels AG, Kroon AA,
the underlying renal parenchyma can be compressed.
de Leeuw PW, van Engelshoven JM. Diagnostic tests
Most often the hematomas resolve over time. On for renal artery stenosis in patients suspected of hav-
occasion, however, the subcapsular fluid collection ing renovascular hypertension: a meta-analysis. Ann
does not resolve. This can lead to altered hemodynam- Intern Med 2001;135:401 – 11.
ics within the compressed parenchyma and a localized [2] Levy JM, Duszak Jr RL, Akins EW, et al. Percutaneous
ischemia. On occasion, similar direct renal compres- transluminal renal angioplasty. American College of
sion may be caused by perirenal fibrosis secondary to Radiology. ACR Appropriateness Criteria. Radiology
perirenal hemorrhage [48,49]. The focal parenchymal 2000;215:1015 – 28.
ischemia activates the renin-angiotensin II pathway [3] Safian RD, Textor SC. Renal-artery stenosis. N Engl J
and hypertension can occur. This process is rare and is Med 2001;344:431 – 42.
[4] Grenier N, Trillaud H. Comparison of imaging meth-
known as a ‘‘Page kidney.’’ Evaluation with either
ods fo renal artery stenosis. BJU Int 2000;86(suppl 1):
renal CT or MR imaging is best for depiction of the 84 – 94.
subcapsular fluid collection and any possible varia- [5] Klassen PS, Svetkey LP. Diagnosis and management of
tions in renal parenchymal enhancement (Fig. 15). renovascular hypertension. Cardiol Rev 2000;8:17 – 29.
Definite management of page kidney includes percu- [6] Angle JF, Hillman BJ. Disorders of renal arterial circu-
taneous or surgical drainage of the hematoma, surgical lation. In: Pollack HM, McClennan BL, editors. Clin-
decapsulation and resection of the hematoma, and ical urography. Philadelphia: WB Saunders; 2000.
partial or total nephrectomy. Renal vein renin sampling p. 2491 – 544.
may help predict the success of surgical intervention if [7] Rimmer JM, Gennari FJ. Atherosclerotic renovascular
the affected side shows hyperreninemia. disease and progressive renal failure. Ann Intern Med
1993;118:712 – 9.
[8] Fenves AZ, Ram CV. Fibromuscular dysplasia of the
Clinical evaluation renal arteries. Curr Hypertens Rep 1999;1:546 – 9.
[9] Lee VS, Rusinek H, Johnson G, Rofsky NM, Krinsky
The role of a clinical evaluation in the detection of GA, Weinreb JC. MR renography with low-dose gado-
renal-mediated hypertension is limited. Certain medi- pentetate dimeglumine: feasibility. Radiology 2001;
cal histories and physical findings can suggest RAS. 221:371 – 9.
A recent history of trauma may provide evidence to [10] Grenier N, Trillaud H, Combe C, et al. Diagnosis of
consider a dissection, posttraumatic stenosis, arterio- renovascular hypertension: feasibility of captopril-sen-
venous communication, or potentially a page kidney. sitized dynamic MR imaging and comparison with cap-
topril scintigraphy. AJR Am J Roentgenol 1996;166:
Flank pain and hematuria can be associated with renal
835 – 43.
tumors. In aortic dissection patients may complain of
[11] Mitty HA, Shapiro RS, Parsons RB, Silberzweig JE.
a pain that radiates to their back. Many of these signs Renovascular hypertension. Radiol Clin North Am
or symptoms may not be present, however, despite 1996;34:1017 – 36.
the underling disease and are of dubious usefulness. [12] Geyskes GG, Oei HY, Puylaert CB, Mees EJ. Renovas-
Laboratory values are also often unreliable. In- cular hypertension identified by captopril-induced
creased serum renin levels may be the result of a changes in the renogram. Hypertension 1987;9:451 – 8.
928 R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929

[13] Harward TR, Poindexter B, Huber TS, Carlton LM, percutaneous transluminal angioplasty for renovascular
Flynn TC, Seeger JM. Selection of patients for renal hypertension: an overview of published series. BMJ
artery repair using captopril testing. Am J Surg 1995; 1990;300:569 – 72.
170:183 – 7. [29] Martin LG, Cork RD, Kaufman SL. Long-term results
[14] Hollenbeck M, Kutkuhn B, Grabensee B. Colour of angioplasty in 110 patients with renal artery steno-
Doppler ultrasound in the diagnosis of transplant renal sis. J Vasc Interv Radiol 1992;3:619 – 26.
artery stenosis. Bildgebung 1994;61:248 – 54. [30] Rees CR, Palmaz JC, Becker GJ, et al. Palmaz stent
[15] Strandness Jr DE. Duplex imaging for the detection of in atherosclerotic stenoses involving the ostia of the
renal artery stenosis. Am J Kidney Dis 1994;24:674 – 8. renal arteries: preliminary report of a multicenter
[16] Olin JW, Piedmonte MR, Young JR, DeAnna S, Grubb study. Radiology 1991;181:507 – 14.
M, Childs MB. The utility of duplex ultrasound scan- [31] Isles CG, Robertson S, Hill D. Management of reno-
ning of the renal arteries for diagnosing significant renal vascular disease: a review of renal artery stenting in ten
artery stenosis. Ann Intern Med 1995;122:833 – 8. studies. QJM 1999;92:159 – 67.
[17] Radermacher J, Chavan A, Schaffer J, et al. Detection [32] Pohl MA. Renal artery stenosis, renal vascular hyper-
of significant renal artery stenosis with color Doppler tension, and ischemic nephropathy. In: Schrier RW,
sonography: combining extrarenal and intrarenal ap- Gottschalk CW, editors. Diseases of the kidney. Bos-
proaches to minimize technical failure. Clin Nephrol ton: Little, Brown; 1997. p. 1367 – 423.
2000;53:333 – 43. [33] Yagura M, Sano I, Akioka H, Hayashi M, Uchida H.
[18] Berland LL, Koslin DB, Routh WD, Keller FS. Renal Usefulness of percutaneous transluminal angioplasty
artery stenosis: prospective evaluation of diagnosis for aortitis syndrome. Arch Intern Med 1984;144:
with color duplex US compared with angiography: 1465 – 8.
work in progress. Radiology 1990;174:421 – 3. [34] Pollard SG, Hornick P, Macfarlane R, Calne RY. Reno-
[19] Baxter GM, Aitchison F, Sheppard D, et al. Colour vascular hypertension in neurofibromatosis. Postgrad
Doppler ultrasound in renal artery stenosis: intrarenal Med J 1989;65:31 – 3.
waveform analysis. Br J Radiol 1996;69:810 – 5. [35] Kalff V, Shapiro B, Lloyd R, et al. The spectrum of
[20] Radermacher J, Chavan A, Bleck J, et al. Use of pheochromocytoma in hypertensive patients with neu-
Doppler ultrasonography to predict the outcome of rofibromatosis. Arch Intern Med 1982;142:2092 – 8.
therapy for renal-artery stenosis. N Engl J Med 2001; [36] Garel L, Dubois J, Robitaille P, et al. Renovascular hy-
344:410 – 7. pertension in children: curability predicted with nega-
[21] Evans AJ, Richardson DB, Tien R, et al. Poststenotic tive intrarenal Doppler US results. Radiology 1995;195:
signal loss in MR angiography: effects of echo time, 401 – 5.
flow compensation, and fractional echo. AJNR Am J [37] Nakhoul F, Green J, Angel A, Ofer A, Ben-Izhak O,
Neuroradiol 1993;14:721 – 9. Lewin M. Renovascular hypertension associated with
[22] Wilman AH, Riederer SJ. Performance of an elliptical neurofibromatosis: two cases and review of the litera-
centric view order for signal enhancement and motion ture. Clin Nephrol 2001;55:322 – 6.
artifact suppression in breath-hold three-dimensional [38] Lewis 3rd VD, Meranze SG, McLean GK, O’Neill Jr
gradient echo imaging. Magn Reson Med 1997;38: JA, Berkowitz HD, Burke DR. The midaortic syn-
793 – 802. drome: diagnosis and treatment. Radiology 1988;167:
[23] Fain SB, King BF, Breen JF, Kruger DG, Riederer SJ. 111 – 3.
High-spatial-resolution contrast-enhanced MR angiog- [39] Lhote F, Guillevin L. Polyarteritis nodosa, microscopic
raphy of the renal arteries: a prospective comparison polyangiitis, and Churg-Strauss syndrome: clinical as-
with digital subtraction angiography. Radiology 2001; pects and treatment. Rheum Dis Clin North Am 1995;
218:481 – 90. 21:911 – 47.
[24] Rubin GD, Dake MD, Napel S, et al. Spiral CT of renal [40] Kawashima A, Sandler CM, Ernst RD, Tamm EP,
artery stenosis: comparison of three-dimensional ren- Goldman SM, Fishman EK. CT evaluation of renovas-
dering techniques. Radiology 1994;190:181 – 9. cular disease. Radiographics 2000;20:1321 – 40.
[25] Johnson PT, Halpern EJ, Kuszyk BS, et al. Renal artery [41] Ridao N, Luno J, Garcia de Vinuesa S, Gomez F,
stenosis: CT angiography – comparison of real-time Tejedor A, Valderrabano F. Prevalence of hyperten-
volume-rendering and maximum intensity projection sion in renal disease. Nephrol Dial Transplant 2001;
algorithms. Radiology 1999;211:337 – 43. 16(suppl 1):70 – 3.
[26] Tegtmeyer CJ, Elson J, Glass TA, et al. Percutaneous [42] Anderson Jr GH, Blakeman N, Streeten DH. The effect
transluminal angioplasty: the treatment of choice for of age on prevalence of secondary forms of hyperten-
renovascular hypertension due to fibromuscular dys- sion in 4429 consecutively referred patients. J Hypertens
plasia. Radiology 1982;143:631 – 7. 1994;12:609 – 15.
[27] Sos TA, Pickering TG, Sniderman K, et al. Percuta- [43] Cooper ME. Pathogenesis, prevention, and treatment
neous transluminal renal angioplasty in renovascular of diabetic nephropathy. Lancet 1998;352:213 – 9.
hypertension due to atheroma or fibromuscular dyspla- [44] Jandeleit-Dahm K, Cooper ME. Hypertension and dia-
sia. N Engl J Med 1983;309:274 – 9. betes. Curr Opin Nephrol Hypertens 2002;11:221 – 8.
[28] Ramsay LE, Waller PC. Blood pressure response to [45] Kenney PJ. Chronic inflammation. In: Pollack HM,
R.P. Hartman et al / Radiol Clin N Am 41 (2003) 909–929 929

Breyer JA, editors. Clinical urography. Philadelphia: [48] Conrad MR, Freedman M, Weiner C, Freeman C, Sand-
WB Saunders; 2000. p. 947 – 76. ers RC. Sonography of the Page kidney. J Urol 1976;
[46] Godley PA, Ataga KI. Renal cell carcinoma. Curr Opin 116:293 – 6.
Oncol 2000;12:260 – 4. [49] McCune TR, Stone WJ, Breyer JA. Page kidney: case
[47] Watson M. Hypertension in polycystic kidney disease. report and review of the literature. Am J Kidney Dis
In: Watson M, Torres V, editors. Polycystic kidney 1991;18:593 – 9.
disease. New York: Oxford University Press; 1996.
p. 407 – 29.
Radiol Clin N Am 41 (2003) 931 – 944

Evaluation of the hypertensive infant: a rational approach


to diagnosis
Christopher G. Roth, MDa, Stephanie E. Spottswood, MDb,*,
James C.M. Chan, MDc, Karl S. Roth, MDd
a
Department of Radiology, Boston Medical Center, Boston University School of Medicine, 88 East Newton Street,
Boston, MA 02118, USA
b
Department of Radiology, Virginia Commonwealth University Health System, 1250 East Marshall Street,
Post Office Box 980615, Richmond, VA 23298 – 0615, USA
c
Department of Radiology, The Barbara Bush Children’s Hospital, The Maine Medical Center,
22 Bramhall Street, Box 14, Portland, ME 04102, USA
d
Department of Pediatrics, Creighton University, 2500 California Place, Omaha, NE 69178, USA

The last half of the twentieth century witnessed an and uses this analysis to provide the basis for a
explosion of technologic advances, which has revolu- rational diagnostic approach to infantile hypertension.
tionized medical care in particular. One area of med-
icine in which technologic applications have led to
major advances is the field of neonatology. Whereas
survival of an infant weighing 750 g in today’s Clinical aspects of diagnosis
intensive care nursery is hardly unusual, in the early
1960s even the son of President John F. Kennedy was One of the chief deterrents to routine blood pres-
unable to survive at twice that weight. Yet, as with all sure measurement in infants has been the irreproduc-
progress, it has come with a price. For example, ibility of results obtained using the inflatable cuff.
the relatively common procedure of umbilical artery This is further complicated by reports that pressure is
catheterization has resulted in an increase in renal affected by waking versus sleeping, abdominal palpa-
artery occlusion. Consequently, renal arterial occlu- tion, sucking and feeding, position, crying, and agita-
sion has assumed a prominent place on the differential tion [2 – 4]. Normal values for blood pressure in
list for renovascular hypertension in infancy [1]. children were defined in the Second Task Force
Although numerous authors have raised the level Report from the National Institutes of Health, pub-
of attention given to detection of hypertension in lished in 1987 [5]. Standards were put forth for
infancy, consensus on a methodologic approach to children under a year and for term infants; the latter
etiology has yet to be reached. As noninvasive or have been corroborated by subsequent reports. Less
minimally invasive imaging techniques continue to well-defined are normative data for prematurely born
improve, it becomes increasingly important to deter- infants, although it is generally agreed that normal
mine which is the most optimal for a given purpose systolic and diastolic pressures are lower than in term
and when in an evaluation it is appropriate. This babies and correlate with body weight and chrono-
article reviews and evaluates the pertinent literature logic age [6 – 8]. Normal pressures tend to increase
from day to day over the first month, further compli-
cating the problem of definition [6 – 8].
* Corresponding author. In the neonate, precise definition of hypertension
E-mail address: sspottsw@hsc.vcu.edu remains controversial, with most authors using the
(S.E. Spottswood). criteria of Adleman [9] delineated from a review of

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00072-1
932 C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944

the existing literature. These include a reproducible


Box 1. Causes of hypertension in the
pressure of greater than 90/60 in a term infant and
neonate
greater than 80/50 in the premature newborn. The
Adleman criteria were defined in 1978, however, and
Renovascular causes
subsequent data show much lower normal pressures
for very small infants who would not likely have
Catheter-associated
survived in 1978. The definition of hypertension for
thromboembolic disease
the preterm baby, according to the Adleman criteria,
Congenital renal artery stenosis
may be a significant overestimate. By contrast, defi-
Mid-aortic coarctation
nition of hypertension in infants younger than
Renal vein thrombosis
12 months takes into account the difficulty of obtain-
Extrinsic renal artery compression
ing reliable diastolic pressures and uses the systolic
(hydronephrosis, hematoma, tumor)
pressure. A systolic pressure above the 95th per-
Fibromuscular dysplasia
centile for age and height as determined by the
Idiopathic arterial calcification
1987 study cited previously [5] taken at least three
Congenital rubella syndrome
times defines an abnormality.
Gruskin et al [10] have noted that morbidity Renal parenchymal and cystic causes
associated with hypertension increases proportionally
with the percentage elevation above the normal in Polycystic kidney disease
adults; using this as a guide, these workers have Multicystic-dysplastic kidney disease
defined a hypertensive crisis in a child as one in Ureteropelvic junction obstruction
which the blood pressure exceeds by 30% the age- Unilateral renal hypoplasia
related norm. The difficulty in using this approach in Congenital nephrotic syndrome
the infant younger than 12 months is the imprecision Tuberous sclerosis
of norms and of measurement of the blood pressure. Acute tubular necrosis
This is especially true of babies born prematurely, for Acute cortical necrosis
whom even the norms are practically difficult to Interstitial nephritis
establish because of the rapid postnatal changes Renal obstruction
taking place. Clearly, this area of definition is left
to the judgment of the individual physician to resolve Miscellaneous causes
on a patient-to-patient basis.
Many causes of neonatal hypertension are, by their Neoplasia
nature, both curable and life threatening (Box 1). Even Neuroblastoma
such causes as renal artery thrombosis, from which Wilms’ tumor
affected infants seem to recover without hypertensive Mesoblastic nephroma
sequelae [11], are intrinsically life threatening and Medication
demand diagnosis. It is the authors’ recommendation Caffeine
that any infant with documented hypertension in the Dexamethasone
first 6 months of life be treated with the respect due Hypercalcemia
any medical and diagnostic emergency. Vitamin D toxicity
Primary (essential) hypertension has not been well Maternal drug addiction
documented to exist in infants and, in any event, must (cocaine, heroin)
be considered a diagnosis of exclusion. The corollary Neurologic
of this is that the chief causes of infantile hyperten- Seizures
sion are secondary, of which approximately 70% are
renovascular [12,13]. The section that follows dis- Cardiovascular, pulmonary, and endocrine
cusses a somewhat controversial issue: more sophis- causes
ticated imaging techniques, which may help to
localize subtle abnormalities of arterial circulation Thoracic aortic coarctation
in the kidney. Although great care is taken to include Intracranial hemorrhage
discussion of all such methodologies available, ad- Bronchopulmonary dysplasia
vancing technology will undoubtedly create new ones Pneumothorax
in the future, which require close scrutiny for their Congenital adrenal hyperplasia
usefulness of application in infants.
C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944 933

Radiologic aspects of diagnosis likely because of the technical difficulties and risks
of anesthesia.
An introduction to renal imaging Among renal parenchymal and cystic diseases
potentially causing infantile hypertension are poly-
The goals of imaging are to detect those infants cystic kidney disease (autosomal-recessive far more
who have renal artery stenosis as the cause of commonly than autosomal-dominant); unilateral renal
hypertension, to predict curability following interven- hypoplasia; congenital nephrotic syndrome; and ac-
tion, and to identify those patients who have renal quired conditions, such as acute tubular necrosis, acute
parenchymal or structural abnormalities as the cause cortical necrosis, and interstitial nephritis. Nonparen-
of their hypertension. The same physiologic and chymal renal causes of hypertension include uretero-
practical considerations that complicate blood pres- pelvic junction obstruction; vesicoureteral junction
sure measurement in infants also challenge the mo- obstruction; and renal obstruction from other causes,
dalities charged with investigating the cause of such as calculi, blood clots, or other mass lesion.
hypertension. The imaging modalities that have been The nonrenal causes of infantile hypertension (see
used in the evaluation of the hypertensive infant Box 1) constitute an array of conditions involving
include intravenous urography, renal scintigraphy, different organ systems including endocrine condi-
ultrasonography, and angiography [14]. CT angiog- tions, such as congenital adrenal hyperplasia; pulmo-
raphy and MR angiography of the renal arteries have nary disorders, such as bronchopulmonary dysplasia
been incorporated into the work-up of the adult and pneumothorax; neoplastic entities, such as Wilms’
hypertensive, but have only anecdotal experience in tumor and neuroblastoma; neurologic conditions,
the infant. such as intracranial hypertension and seizures; and
miscellaneous causes, such as total parenteral nutri-
Etiologic considerations in imaging tion, hypercalcemia, adrenal hemorrhage, and medi-
cations including dexamethasone, adrenergic agents,
Fortunately, the history and physical examination and others [1]. Most of the nonrenal causes can be
frequently suggest the underlying cause of hyperten- suggested by the history, physical examination, and
sion in the infant. The potential causes are numerous laboratory analysis.
and the recognized imaging modalities, which are
variably invasive, often yield mutually exclusive data. Application of imaging to diagnosis of renal
Most cases of hypertension in infants (see Box 1) parenchymal disease
are caused by renovascular, renal parenchymal, or
cystic disease [1]. Renovascular disorders accounted Sonography
for 48% of neonatal hypertension in a recent study Because most cases of infantile hypertension are
[15]. Catheter-associated thromboembolic disease is caused by renal abnormalities, a diagnostic approach
the most common offender in this category. The focused on the kidneys is vital. Renal sonography is
mechanism is believed to be disruption of the vascular typically used as the initial imaging modality in the
endothelium of the umbilical artery following catheter evaluation of the hypertensive infant because of its
line placement, which initiates thrombus formation. convenience, accessibility, noninvasiveness, and lack
This may propagate directly or embolize to the renal of radiation exposure. It is highly sensitive in detec-
artery causing regions of ischemia or infarction with tion of many of the parenchymal diseases of the
increased renin release. Other renovascular etiologies kidney (see Box 1), and for evaluating anomalies of
include congenital renal artery stenosis, mid-aortic the renal collecting system. Sonography has replaced
coarctation, renal vein thrombosis, and fibromuscular the intravenous urogram as the initial imaging mo-
dysplasia. Finally, extrinsic compression of the renal dality in the evaluation of infants and small children
artery can result from hydronephrosis; tumor; or with hypertension [20]. Sonography is comparable
hematoma (eg, from adrenal hemorrhage) [16]. Renal with intravenous urography in the assessment of renal
arteriography is the gold standard for the diagnosis of size and hydronephrosis, without the risks of intra-
renovascular disease in the adult. Treatment also may venous contrast administration and patient exposure
be offered by angiography, because limited data in to ionizing radiation.
the older pediatric population have shown that per- Sonographic evaluation of the kidneys is per-
cutaneous transluminal angioplasty can effectively formed with a combination of gray-scale, color Dopp-
treat renovascular hypertension [17 – 19]. As is dis- ler, and duplex Doppler imaging. Gray-scale imaging
cussed, however, angiography is less often performed depicts a structural rendition of the kidney based on
in the evaluation of neonatal hypertension, most acoustic interfaces (acoustic impedance differences
934 C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944

between adjacent tissues). Its primary use is for


anatomic detail. Color Doppler imaging superim-
poses a color-coded velocity flow scale, based on
the frequency shift of moving tissues, onto the gray-
scale image. Duplex Doppler imaging provides a
spectral trace recording frequency changes over time,
reflecting the velocity profile. The Doppler modalities
are useful for evaluation of vascular structures.

Gray-scale sonography. Gray-scale sonography is


initially used to assess the kidneys for any paren-
chymal or structural abnormality. Coronal, sagittal,
and transverse imaging of the kidneys is performed
with a high-resolution transducer to simulate a three-
dimensional view of the renal parenchyma and collect-
ing system. The collecting system is evaluated for Fig. 1. Normal renal sonogram of 9-day-old infant with
hydronephrosis, which can result from ureteropelvic hypertension. Sagittal image of the right kidney demon-
junction obstruction, ureterovesicular junction ob- strates normal parenchymal echogenicity with good cortico-
struction, bladder outlet obstruction, or vesicoureteral medullary differentiation. Note normal, triangular-shaped,
reflux. Hydronephrosis is easily perceived sonograph- hypoechoic renal pyramids. Duplex Doppler examination
ically as dilatation of the renal collecting system. was normal.
Hydronephrosis without hydroureter is typical of con-
genital ureteropelvic junction obstruction; hydro- interfaces produce exceptionally bright kidneys (in-
nephrosis with hydroureter is apparent with creased echogenicity) with ultrasound evaluation. Au-
ureterovesicular junction obstruction or vesicoureteral tosomal-dominant polycystic kidney disease, which is
reflux. Renal size is assessed, and any asymmetry in less common at this age, exhibits macroscopically
length greater than 5 mm may indicate unilateral renal visible cysts of varying size. Mesoblastic nephroma,
disease. Normal renal length in a full-term neonate often diagnosed in infancy, and Wilms’ tumor, usually
ranges from 4 to 5.5 cm [21]. In the neonate and young diagnosed in early childhood, manifest sonographi-
infant, the renal parenchyma demonstrates increased cally as a mass arising from the kidney. Wilms’ tumor
cortical echogenicity because the glomeruli occupy a may be accompanied by tumor invasion of the renal
larger volume of the cortex in infants (18%) as vein, which can also be detected sonographically.
compared with older children and adults (8.6%), and Hypertension can occur as a result of increased renin
20% of the loops of Henle are located within the cortex production by tumor cells [23].
rather than within the medulla [22]. Increased numbers In the infant with renal artery thrombosis, there is
of anatomic structures in the cortex create an increased little parenchymal abnormality in the acute phase of
number of interfaces for the ultrasound beam to vascular obstruction, but with time there is loss of
contact, resulting in increased cortical echogenicity. corticomedullary differentiation with diffusely in-
Additionally, there is a relatively larger volume of creased echogenicity, and decreased renal size, indi-
medulla in the neonatal kidney than in the adult kidney, cating chronic ischemia. Chronically ischemic or
with cortico-medullary ratio of 1.64:1 in the neonate infarcted kidneys appear markedly shrunken and
and 2.59:1 in the adult [22]. This results in a striking abnormally echogenic (Fig. 2). Renal vein thrombosis
corticomedullary differentiation not seen in older chil- likewise exhibits poor corticomedullary differentia-
dren and adults (Fig. 1). Loss of this corticomedullary tion, but the affected kidney is enlarged. Thrombus,
differentiation in the neonate reflects diffuse renal usually manifested by intraluminal echogenic ma-
disease or congenital dysplasia. terial, may be detected in the renal artery or vein,
Renal cystic diseases are clearly depicted by or in the abdominal aorta. Intraluminal thrombus,
sonography. Autosomal-recessive polycystic kidney however, occasionally appears anechoic (without
disease characteristically reveals bilaterally enlarged echoes, indistinguishable from the patent blood ves-
kidneys with diffusely and uniformly increased echo- sel lumen) and color Doppler imaging is required to
genicity and loss of the normal corticomedullary demonstrate its presence.
differentiation. The individual cysts, which actually Although the presence of echogenic thrombus
represent dilated collecting ducts, are too small to be within the lumen of the aorta or the renal artery is
resolved sonographically, but their numerous wall highly suggestive of thrombus, a recent study of
C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944 935

renal vascular imaging, the abdominal aorta can be


examined from the diaphragm to the bifurcation in
the coronal plane, to look for disruption of color flow
by thrombus. Color imaging also is useful in distin-
guishing between anechoic renal hilar vascular struc-
tures from a similar-appearing dilated ureter or
collecting system.
The renal vasculature can be evaluated best sono-
graphically by duplex Doppler imaging. Various
quantitative and qualitative measures have been
designed to define renovascular disease. The classic
duplex Doppler findings in arterial stenosis are an
increase in blood flow velocity and spectral broad-
Fig. 2. Abnormal renal sonogram in young child with
renovascular hypertension. Sagittal sonogram reveals loss of ening. Spectral broadening denotes a widening in the
corticomedullary differentiation (compare with Fig. 1) and spectrum of detected velocities, which is a manifes-
focal areas of chronic cortical scarring (arrows). Note focal tation of turbulent flow through a stenotic segment.
upper pole caliectasis (white arrow). Because renal blood flow is parabolic and the spec-
trum is inherently widened, however, spectral broad-
infantile hypertension has demonstrated no causal ening is not a valid means of defining turbulent flow
relationship between the identification of renal or in the renal arteries [28].
aortic thrombus and renovascular hypertension [15]. Other objective measurements have been designed
Conversely, many normotensive patients fulfilled to define sonographically renovascular hypertension.
gray-scale sonographic criteria for thromboembolism. The acceleration index and resistive index have been
A prospective study using ultrasound to detect aortic used to identify renovascular hypertension [28]. The
thrombus was positive in 12 of 71 patients in the acceleration index is determined by the intersection
neonatal intensive care unit; only one of these pa- of a line indicating the upstroke of systole with a line
tients developed hypertension and two normotensive drawn 1 second later perpendicular to the baseline;
patients subsequently proved to have aortic thrombus the height of this line is divided by the ultrasound
did not have sonographically detectable renal artery frequency. The resistive index, a more commonly
thrombus [24]. used measurement, is the ratio of peak diastolic
The reported incidence of catheter-associated velocity to peak systolic velocity (Fig. 3). Patriquin
thromboembolism in infants is highly variable, rang- et al [28] studied 20 children in whom renal artery
ing from 3.5% to 23% in autopsy series to 95% in stenosis was suspected. Doppler tracings from at least
prospective ultrasound studies [25]. Clearly, it is a three segmental or intralobar arteries were obtained in
common complication, and the presence of echogenic
intravascular material associated with systemic hyper-
tension is highly suggestive of renovascular disease;
however, earlier reports have demonstrated that gray-
scale imaging alone does not identify all cases of
renovascular hypertension. If the affected renal ves-
sels are beyond first-order branch vessels that cannot
be resolved reliably sonographically, and there are no
associated morphologic changes in the renal paren-
chyma, there is no gray-scale sonographic abnor-
mality. It is postulated that small-vessel renal disease
can be identified on gray-scale imaging as a dotted
corticomedullary junction [26].

Color Doppler imaging. Color Doppler imaging


Fig. 3. Spectral Doppler image in 1-year-old child with renal
can be used as an adjunct to gray-scale imaging in artery stenosis. Resistive index (RI) is measured (electronic
the detection of intraluminal thrombus, which may be cursors) as the ratio of the peak diastolic velocity to the peak
isoechoic to flowing blood. Color Doppler imaging systolic velocity (RI = 1  [D/S]). In this case 1  (28.8/
can show absent flow distal to thrombus and the 83.5) = 0.66. Note normal Doppler waveform from the
presence of collateral vessels [27]. In addition to renal artery.
936 C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944

each patient and the acceleration index and resistive


index calculated. Both indices were significantly
lower in stenotic arteries (the acceleration index to
a greater extent), with clear discrimination between
normal arteries and those with at least a 75% angiog-
raphic stenosis.
Normal renal arteries were associated with an
acceleration index of 4 to 7; renal arteries with at
least 75% stenosis ranged from 0.7 to 2.6. Although
used more commonly in practice, the resistive index
varied less with renal arterial stenosis with a resistive
index of 0.56 or less predicting stenosis with 95%
probability [28].
Conversely, a prospective study of hypertensive
children aged 12 days to 15 years defined a subset of
angiographically proved renovascular hypertensive
patients with negative Doppler examinations. The
Doppler ultrasound examinations, however, were
assessed qualitatively [29]. Specifically, they used
pattern recognition of the tardus-parvus phenomenon
[30], where pulsus tardus is the slowed, delayed
systolic upsweep, and pulsus parvus represents a
dampened maximal systolic peak, characteristic of a
severe stenosis (Fig. 4). The presence of multiple
renal arteries and segmental lesions accounted for
most false-negative Doppler examinations. In this
series, hypertensive patients with a negative duplex Fig. 5. Renal arteriogram in a 3-year-old patient with
Doppler examination generally had vascular lesions uncontrollable hypertension and history of neurofibroma-
amenable to endovascular or surgical treatment with a tosis. Middle aortic syndrome with renal artery stenosis.
high rate of success. It was concluded that for these Aortic arteriogram reveals marked, long-segment stenosis
reasons Doppler sonography may be unreliable in the and irregularity of the aorta extending from the suprarenal
evaluation of renal artery stenosis. The authors sug- region to just above the bifurcation. The right renal artery is
occluded at its origin (curved arrow) and the left renal artery
gested that with a negative Doppler sonogram and a
(straight arrow) is markedly stenotic. Note also occlusion of
the hepatic artery (large arrow). There is marked enlargement
of the inferior mesenteric artery and left colic artery
(arrowheads), and multiple lumbar arteries. (Courtesy of
Jaime Tisnado, Medical College of Virginia, Richmond, VA.)

strong suspicion for renovascular hypertension, selec-


tive or superselective arteriography in association
with segmental venous renin sampling should be
performed because an angiographically demonstrated
causal lesion, if treated, most likely results in cure
[29]. Angiography is generally deferred in the
neonate, however, and medical treatment (often with
angiotensin converting enzyme inhibitors [ACEI]) is
the mainstay.
The aforementioned studies using Doppler ultra-
Fig. 4. Renal sonogram with spectral Doppler image in
sound included very few infants. Technical factors,
1-year-old child with renal artery stenosis demonstrating
tardus-parvus phenomenon. Duplex Doppler spectral tracing
including the inherent difficulty in obtaining Doppler
of the left renal artery reveals delayed and dampened systolic signal from multiple intrarenal vessels, the long
upsweep (arrow) typical of renal artery stenosis (note duration of the examination, and the frequent lack
continuous venous waveform below the line). Compare with of visualization of the proximal renal vasculature
normal renal arterial waveform in Fig. 3. because of bowel gas, challenge the implementation
C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944 937

of this technique at all, let alone in infants. Multiple nique in the intensive care setting. In the persist-
additional factors further complicate the application ently hypertensive neonate in which duplex Doppler
of such techniques in the neonatal ICU, including the sonography is not technically feasible, the identifica-
use of portable machines; the presence of life support tion of intraluminal thrombus on gray-scale imaging
lines and tubes; and the likelihood that the patient is suggests the diagnosis. The absence of intraluminal
dependent on mechanical ventilation, which renders thrombus on gray-scale imaging, however, does not
duplex Doppler sonography virtually impossible (es- exclude renovascular hypertension and further diag-
pecially in the setting of high-frequency ventilation). nostic investigation should be pursued.
Sonography is a versatile modality that offers
several parameters for evaluating the hypertensive Angiography
infant. The implementation of duplex Doppler sonog- The accuracy of Doppler sonography in the diag-
raphy is limited by operator skill and experience and nosis of renal artery stenosis has been compared with
by the technical difficulties in performing this tech- renal angiography, which is considered the gold

Fig. 6. Technetium (Tc) 99m MAG3 ACEI renogram in a 13-day-old infant with unexplained hypertension. (A, B) Normal
baseline study. Posterior images of the kidneys were obtained following the intravenous administration of Tc 99m MAG3 (initial
flow images were unremarkable) (A). Note symmetric uptake and excretion of tracer, followed by visualization of the urinary
bladder. Normal time-activity curve demonstrates peak renal activity at 3 minutes (normal) and a differential function of 48.8%
(left kidney) and 51.2% (right kidney) (B). (C, D) Normal enalaprilat study. Posterior images of the kidneys were obtained
following the intravenous administration of enalaprilat, followed by intravenous Tc 99m MAG3 (C). Peak renal activity on the
time activity curves is demonstrated at 2 minutes (normal) (D). There is normal differential function: 45% (left kidney) and 55%
(right kidney), and no significant renal cortical retention of tracer.
938 C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944

Fig. 6 (continued ).

standard in the evaluation of renovascular hyperten- essentially abandoned in the evaluation of the hyper-
sion [14,15,20]. Despite the high incidence of reno- tensive infant [20].
vascular hypertension in children relative to adults, The potential benefit of arteriography in the
the use of renal arteriography in children has been evaluation of the pediatric hypertensive patient is the
limited. The necessity for general anesthesia generally opportunity for definitive treatment. Percutaneous
preempts the use of conventional arteriography in transluminal angioplasty has been demonstrated to
infants. Catheter-related vascular injury and radiation be effective in the adult population and has recently
exposure are other potential adverse considerations. been applied to the pediatric population with success
Although intra-arterial digital subtraction arteriogra- [17,18]. Nonetheless, virtually no data are available
phy requires substantially less intravascular contrast regarding the use of percutaneous transluminal angio-
material and shortens the duration of the procedure plasty in infants. The highest rate of success in
compared with traditional arteriography, resulting in children has been associated with nonostial, short-
less radiation exposure, it has not substantially in- segment main renal arterial lesions; the technical
creased the use of arteriography in the diagnosis of difficulty in traversing ostial lesions, with or without
infantile hypertension. When performed, digital sub- aortic involvement, often precludes successful angio-
traction arteriography images may be compromised by plasty [32].
the presence of bowel gas, although both intravenous Most angiographically demonstrated lesions in
glucagon and abdominal compression can mitigate this cases of pediatric hypertension are related to intrinsic
problem [31]. Intravenous digital subtraction arteriog- vascular disorders, such as fibromuscular dysplasia,
raphy has been attempted as an alternative to arteriog- neurofibromatosis, and other undifferentiated vascu-
raphy in the evaluation of renovascular hypertension litides. During arteriography, pharmacologic maneu-
with limited success. Intravenous digital subtraction vers, such as epinephrine infusion, can determine the
arteriography requires a higher contrast load and the hemodynamic significance of renal arterial lesions.
vessels of interest are frequently not well opacified by One of the inherent advantages of angiography is that
this technique. Lesions beyond the first-order branch the main renal artery, and the intrarenal segmental,
vessels are not demonstrated by intravenous digital subsegmental, and any accessory renal arteries are well
subtraction arteriography and this technique has been demonstrated and any of these vessels may be affected
C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944 939

Fig. 6 (continued ).

in the previously mentioned disorders (Fig. 5). Al- with medical therapy and not with percutaneous
though these entities do occur in infants, they are not transluminal angioplasty, which argues against the
typically the chief diagnostic considerations in this age implementation of arteriography [1,15,33]. Because
group, especially in neonatal patients. the usual first-line therapy in neonatal renovascular
In infants, the most common underlying abnor- hypertension is ACEI and revascularization is gen-
mality is catheter-related thromboembolism. Aortic erally not an option, it is more important to identify
and renal arterial thrombus can be demonstrated with cases of bilateral renal ischemia and renal ische-
contrast aortography and renal arteriography per- mia in a solitary kidney in which ACEI therapy
formed by the offending umbilical artery catheter is contraindicated. Renal scintigraphy is the least
[25]. Many studies have proved the efficacy of invasive and most reliable means of providing
arteriography in documenting the presence of throm- this information.
bus in association with umbilical arterial cathe-
terization. There has been very poor correlation, Renal scintigraphy
however, between the presence of thrombus and Renal scintigraphy can yield valuable functional
clinical signs and symptoms. The variably reported data with variable anatomic detail. In the presence of
incidence of arterial thrombus associated with um- unilateral renal artery stenosis, conventional radio-
bilical artery catheterization is high enough that it nuclide scintigraphy may show evidence of relatively
may be an incidental finding in some instances. In diminished renal perfusion and function of the affect-
any event, catheter-associated aortic and renal arte- ed kidney. Because of the autoregulatory mechanism,
rial thrombus has been treated relatively successfully however, mediated by the renin-angiotensin system,
940 C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944

Fig. 6 (continued ).

the glomerular filtration rate (GFR) may be main- If the baseline study is normal, the administration
tained at a normal level and the scintigram may be of the ACEI eliminates the renin-angiotensin compen-
normal. Performing the examination in conjunction sation and thereby decreases the renal perfusion
with an ACEI greatly increases the sensitivity and commensurate with the degree of stenosis. This trans-
specificity of renal scintigraphy for detecting hemo- lates to a decrease in function in the well-compensated
dynamically significant renal artery stenosis; the kidney. There is high probability of hemodynami-
sensitivity and specificity are each approximately cally significant renal artery stenosis when there is
90% [34]. (1) marked change in the renogram curve, (2) unilat-
When renal arterial stenosis reaches 60% of the erally reduced relative uptake of tracer, or (3) unilat-
cross-sectional diameter of the artery, the kidney erally prolonged renal and parenchymal transit time.
responds by increasing its output of renin, stimulating In cases of very severe renal artery stenosis (up to
production of angiotensin II, which augments falling 95%) there is no significant change from baseline after
GFR by increasing tone in the efferent arterioles at ACEI administration with at most minimal residual
the cost of generalized vasoconstriction, resulting in renal function. When renal arterial stenosis has
systemic hypertension [14]. The administration of an resulted in complete obstruction, the baseline scinti-
ACEI blocks the production of angiotensin II, which gram may demonstrate some blood pool activity
decompensates renal function. caused by collateral vessels and there is no change
An ACEI scintigraphy capitalizes on this physio- after ACEI.
logic compensation mechanism. A baseline renogram Renal scintigraphy for the evaluation of renal artery
is first performed, which may be normal with a renal stenosis can be performed with a choice of ACEI:
arterial stenosis of up to 70% to 80% [14]. Beyond captopril or enalaprilat. Enalaprilat is administered
this range, renin-angiotensin compensation may be intravenously, and unlike orally administered capto-
incomplete and the baseline study may show dimin- pril, its pharmacologic effect is not dependent on rate
ished function. If the kidney has infarcted and there is of gastrointestinal absorption. ACEIs can cause sig-
no residual function, the baseline study results in nificant hypotension; blood pressure and heart rate are
nonvisualization of the involved kidney. monitored before and during ACEI administration.
C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944 941

The baseline and follow-up ACEI scintigraphy The disposition of glomerular agents in the kidney
can be performed with either glomerular or tubular is dependent on the GFR; the rate of accumulation of
radiopharmaceuticals. The original studies performed radiotracer is directly proportional to the GFR. The
in pediatric patients used a glomerular agent, Tc 99m rate of glomerular agent accumulation can be
diethylenetriamine pentaacetic acid (DTPA) with expressed as the slope of the curve of computer-
captopril [35]. Subsequently, Tc 99m mercaptoacetyl- generated graphs at fixed intervals and differential
triglycine (MAG3), a tubular agent, has been used for renal function of each kidney can be derived (Fig. 6).
ACEI renography. It is preferred over Tc 99m DTPA With Tc 99m DTPA, the scintigraphic manifestation
in patients with elevated serum creatinine, because of of decreased renal function following ACEI adminis-
its higher renal extraction. tration is decreased extraction and delayed appearance

Fig. 7. Tc 99m MAG3 renogram. Renal artery stenosis in an older child with hypertension. (A) Posterior images of the kidneys
were obtained following the intravenous administration of enalaprilat (initial flow images were unremarkable). The early images
reveal reduced tracer uptake and function of the relatively smaller right kidney, whereas the normal left kidney reveals normal
accumulation and ureteral excretion of tracer. The delayed images demonstrate marked retention of tracer in right kidney (arrow),
consistent with hemodynamically significant renal artery stenosis. (B) Normal baseline Tc 99m MAG3 renogram. (Courtesy of
Massoud Majd, Children’s National Medical Center, Washington, DC.)
942 C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944

of the radiotracer in the collecting system. The affected tion, and measurements of residual cortical activity
kidney demonstrates relatively decreased uptake. can be displayed graphically (see Fig. 7).
Tubular agents are secreted by the proximal In hypertensive neonates without an umbilical
tubules, a function that is maintained in the setting arterial catheter, an abnormal ACEI study indicates
of a falling GFR. As urine production in the ischemic renal artery stenosis. A more common cause of
kidney decreases after ACEI administration, the tu- hypertension in neonates, however, usually transient,
bular agent accumulates and remains in the cortex is narrowing of the renal artery because of thrombosis
because of the fall in urine production. The affected as a complication of the umbilical artery catheter
kidney demonstrates parenchymal retention of tracer (Fig. 8). The value of ACEI renography in these
(Fig. 7). Tc 99m MAG3 has an advantage over Tc neonates is to determine whether it is safe to treat
99m DTPA in that the images are of higher resolu- them with ACEI therapy.

Fig. 8. Tc 99m MAG3 renogram. Renal artery thrombosis. Neonate who became hypertensive a few days following umbilical
artery catheter placement. (A) Initial posterior images obtained following administration of enalaprilat reveal a normal-appearing
right kidney, and a smaller, irregularly contoured left kidney, presumably developmental. Delayed images reveal normal
excretion from the left kidney, but marked retention of MAG3 in the right kidney (arrow) caused by partial obstruction of the
renal artery. Time-activity curves generated from region of interest drawn around the right kidney demonstrate (B) normal pre-
ACEI function of the right kidney (pre-captopril) and (C) impaired post-ACEI function (post-enalaprilat). (Courtesy of Massoud
Majd, Children’s National Medical Center, Washington, DC.)
C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944 943

In addition to its high sensitivity and specificity sion in the young infant. The numerous etiologies of
for hemodynamically significant renal artery stenosis, hypertension have been discussed and normative
a major benefit of ACEI renography is that a positive blood pressure data for neonates and infants have
study indicates a high probability that blood pressure been provided. Techniques for accurate blood pres-
is reduced following angiographic intervention [31]. sure measurement in the intensive care setting and for
Although this procedure has been performed safely in routine outpatient settings, are discussed.
young children [18], subsequent intervention with The lengthy discussion of radiologic approach
percutaneous transluminal angioplasty is an unlikely to imaging can be summarized with the following
consideration in the young infant. suggested algorithm. Initial screening should be per-
formed with gray-scale sonography, to identify renal
parenchymal or collecting system abnormalities, in-
Future considerations for imaging diagnosis cluding mass lesions and congenital anomalies.
Further imaging with color and duplex Doppler
CT angiography of the renal arteries has had sonography detects renal arterial or aortic thrombosis,
promising results in the detection of renal artery and alterations in the arterial waveform caused by
stenosis in the adult population. New developments intrinsic or extrinsic renal artery narrowing. The major
in CT technology, including spiral CT and multidetec- limitation of Doppler sonography is the recognition
tor CT, allow volumetric acquisitions during a single that disease in accessory renal arteries or in small
breathhold. The volume of acquired data can then be segmental intrarenal arteries may frequently be unde-
reformatted for display in any plane. Sensitivity and tected. Functional imaging with ACEI renography
specificity for detection of hemodynamically signifi- should follow renal sonography to detect hemo-
cant renal artery stenosis in the adult population have dynamically significant renovascular disease (with a
been as high as 92% and 83%, respectively [36], and sensitivity and specificity of approximately 90%);
90% and 97%, respectively [37]. High sensitivity and intravenous enalaprilat is the preferred ACEI.
specificity for this modality may be caused by the Angiography should be reserved for older children
ostial location of stenotic lesions seen in adults. Al- in whom interventional percutaneous angioplasty may
though little data are currently available describing use be more feasible. A young infant with hypertension
of CT angiography in infants and children, potential caused by renal artery stenosis should be controlled
advantages include relative speed of image acquisition medically until he or she is large enough to undergo
(which may obviate need for sedation), and minimal angiography and angioplasty successfully. CT angiog-
invasiveness, as compared with angiography. Potential raphy and MR angiography, although promising in the
disadvantages include inability to breathhold, and adult population, may not adequately resolve the small
limitations in the evaluation of small accessory, seg- intrarenal vessels, which are frequently the culprit in
mental, or intrarenal arteries [37], which unfortunately renovascular hypertension of infancy.
are frequently involved in infants with renal artery
stenosis. Additionally, larger doses of intravenous
contrast are required with CT angiography than with
References
intra-arterial digital subtraction arteriography [37].
As with CT angiography, little data are available
[1] Flynn JT. Neonatal hypertension: diagnosis and treat-
on the use of MR angiography of the renal arteries in ment. Pediatr Nephrol 2000;14:332 – 41.
infants. Anecdotally, MR angiography has been [2] Lee YH, Rosner B, Gould JB, et al. Familial aggrega-
attempted by one of the authors, in an infant with tion of blood pressure of newborn infants and their
intractable hypertension, and in an older child with mothers. Pediatrics 1976;58:722 – 9.
middle aortic syndrome. Limitations were difficulty [3] Sinkin RA, Phillips BL, Adelman RD. Elevation in
resolving the small renal arteries, motion artifact systemic blood pressure in the neonate during abdomi-
during imaging, and the need for sedation for this nal examination. Pediatrics 1985;76:970 – 2.
relatively long study. Obvious advantages include [4] Moss AJ, Duffie Jr ER, Emmanouilides G. Blood pres-
lack of ionizing radiation or intravenous contrast. sure and vasomotor reflexes in the newborn infant.
Pediatrics 1963;32:175 – 9.
[5] The National Heart, Lung, and Blood Institute (Bethes-
da, MD). Report of the Second Task Force on Blood
Summary Pressure Control in Children. Pediatrics 1987;79:1 – 25.
[6] Tan KL. Blood pressure in very low birth weight in-
This article reviews the literature and describes a fants in the first 70 days of life. J Pediatr 1988;112:
methodologic approach to the diagnosis of hyperten- 266 – 70.
944 C.G. Roth et al / Radiol Clin N Am 41 (2003) 931–944

[7] McGarvey ST, Zinner SH. Blood pressure in infancy. [23] Cohen MD. Imaging of children with cancer. St. Louis:
Semin Nephrol 1989;9:260 – 6. Mosby Year Book; 1992.
[8] Zubrow AB, Hulman S, Kushner H, et al. Determi- [24] Oppenheimer DA, Carroll BA, Garth KE. Ultrasonic
nants of blood pressure in infants admitted to neonatal detection of complications following umbilical arterial
intensive care units: a prospective multicenter study. catheterization in the neonate. Radiology 1982;145:
J Perinatol 1995;15:470 – 9. 667 – 72.
[9] Adelman RD. Neonatal hypertension. Pediatr Clin [25] Ford KT, Teplick SK, Clark RE. Renal artery embo-
North Am 1978;25:99 – 110. lism causing neonatal hypertension. Radiology 1974;
[10] Gruskin AB, Lerner GR, Fleischmann LE. Manage- 113:169 – 70.
ment of acute and primary hypertension in children. [26] Garel LA, Pariente DM, Gubler MC, et al. The dotted
In: Murakami K, Kitagawa T, Yabuta K, et al, editors. cortico-medullary junction: a sonographic indicator of
Recent advances in pediatric nephrology. Amsterdam: small vessel disease in hypertensive children. Radiology
Elsevier; 1987. p. 121 – 6. 1984;152:419 – 22.
[11] Adelman RD. Long-term follow-up of neonatal reno- [27] Deeg KH, Wolfel D, Rupprecht TH. Diagnosis of neo-
vascular hypertension. Pediatr Nephrol 1987;1:35 – 41. natal aortic thrombosis by colour coded Doppler so-
[12] Inglefinger J. Nephrology forum: renovascular hyper- nography. Pediatr Radiol 1992;2:62 – 3.
tension in children. Kidney Int 1993;43:493 – 505. [28] Patriquin HB, Lafortune M, Jequier J-C, O’Regan S,
[13] Singh HP, Hurley RM, Myers TF. Neonatal hyperten- Garel L, Landriault J, et al. Stenosis of the renal artery:
sion: incidence and risk factors. Am J Hypertens 1992; assessment of slowed systole in the downstream circu-
5:51 – 5. lation with Doppler sonography. Radiology 1992;184:
[14] Sfakianakis GN, Bourgoignie JJ, Georgiou M, Guerra 479 – 85.
JJ. Diagnosis of renovascular hypertension with ACE [29] Garel L, Dubois J, Robitaille P, Russo P, Filiatrault D,
inhibition scintigraphy. Radiol Clin North Am 1993; Grignon A, et al. Renovascular hypertension in chil-
31:831 – 48. dren: curability predicted with negative intrarenal
[15] Chandar JJ, Sfakianakis GN, Zilleruelo GE, Guerra JJ, Doppler UAS results. Radiology 1995;195:401 – 5.
Georgiou MF, Abithol CL, et al. ACE inhibition scin- [30] Kotval PS. Doppler waveform parvus and tardus.
tigraphy in the management of hypertension in chil- J Ultrasound Med 1989;8:435 – 40.
dren. Pediatr Nephrol 1999;13:493 – 500. [31] Tonkin IL, Stapleton FB, Roy S. Digital subtraction
[16] Starinsky R, Manor A, Segal M. Non-functioning kid- angiography in the evaluation of renal vascular hyper-
ney associated with neonatal adrenal hemorrhage: report tension in children. Pediatrics 1988;81:150 – 7.
of two cases. Pediatr Radiol 1986;16:427 – 9. [32] Mali WP, Puijlaert AJ, Kouwenberg HJ, Klinge J,
[17] Tyagi S, Kaul UA, Satsangi DK, Arora R. Percutane- Donckerwolcke RA, Geijskes BG, et al. Percutaneous
ous transluminal angioplasty for renovascular hyper- transluminal renal angioplasty in children and adoles-
tension in children: initial and long-term results. cents. Radiology 1987;165:391 – 4.
Pediatrics 1997;99:44 – 9. [33] Caplan MS, Cohn RA, Langman CB, Conway JA,
[18] Courtel VC, Soto B, Niaudet P, Gagnadoux MF, Car- Shkolnik A, Brouillette RT. Favorable outcome of neo-
teret M, Quignodon JF, et al. Percutaneous translumi- natal aortic thrombosis and renovascular hypertension.
nal angioplasty of renal artery stenosis in children. J Pediatr 1989;115:291 – 5.
Pediatr Radiol 1998;28:59 – 63. [34] Taylor AT, Fletcher JW, Nally JV, et al. Procedure for
[19] Stanley P, Hieshima G, Mehringer M. Percutaneous diagnosis of renovascular hypertension. J Nucl Med
transluminal angioplasty for pediatric renovascular hy- 1998;39:1297 – 302.
pertension. Radiology 1984;153:101 – 4. [35] Majd M, Potter BM, Guzzetta PC, et al. Effect of
[20] Zerin JM, Hernandez RJ. Renal imaging in children with captopril on efficacy of renal scintigraphy in detection
persistent hypertension. Pediatr Clin North Am 1993; of renal artery stenosis. J Nucl Med 1983;24:23.
40:165 – 78. [36] Rubin GD, Dake MD, Napel S. Spiral CT of renal
[21] Rosenbaum DM, Korngold E, Teele RL. Sonographic artery stenosis: comparison of three-dimensional ren-
assessment of renal length in normal children. Am J dering techniques. Radiology 1994;190:181 – 9.
Radiol 1984;142:467 – 9. [37] Kim TS, Chung JW, Park JH. Renal artery evaluation:
[22] Hricak H, Slovis TL, Callen CW, et al. Neonatal comparison of spiral CT angiography to intra-arterial
kidneys: sonographic anatomic correlation. Radiology DSA. J Vasc Interv Radiol 1998;9:553 – 9.
1983;147:699 – 702.
Radiol Clin N Am 41 (2003) 945 – 961

CT urography and MR urography


Akira Kawashima, PhD, MD*, James F. Glockner, MD, Bernard F. King, Jr, MD
Department of Radiology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA

The uroradiologic evaluation of patients with CT imaging has evolved from single-detector into
common and complex disease is changing rapidly. multidetector row helical volumetric acquisition tech-
Intravenous (excretory) urography has been the initial niques, and these advances have had a significant
evaluation for upper tract imaging in patients with impact on imaging of the urinary tract. Application of
hematuria, flank pain, and other urologic diseases for multidetector row helical volumetric CT evaluation of
the past three decades [1,2]. The imaging investiga- the urinary tract has been termed ‘‘CT urography.’’
tion of hematuria usually began with an abdominal The concept of CT urography is attractive because
radiograph for the detection of opaque urinary calculi. both the renal parenchyma and urothelium can be
Since Smith et al [3] demonstrated the value of evaluated at a single comprehensive examination.
unenhanced CT for the evaluation of patients with This primary use of CT urography potentially allows
acute flank pain in comparison with intravenous shortening the duration of the diagnostic evaluation in
urography in 1995, in many centers intravenous many patients. CT urography is becoming the defin-
urography has been replaced by unenhanced CT for itive study for patients with hematuria.
evaluation of patients with suspected ureteral calculi. Another alternative to conventional imaging of the
The remaining major indication for intravenous urog- urinary tract is MR urography. MR urography using
raphy is hematuria. either heavily T2-weighted pulse sequences or gado-
Patients with hematuria require evaluation of both linium-enhanced T1-weighted sequences has shown
the renal parenchyma and the urothelium. Intra- potential to detect, localize, and characterize collect-
venous urography remains the initial imaging modal- ing system abnormalities. Because neither iodinated
ity of choice for assessing the upper urinary tract. The intravenous contrast nor ionizing radiation is used, it
limitations of intravenous urography are frequently is safe in patients with contraindication to iodinated
complemented with the supplemental use of ultra- contrast media, in young patients, and in pregnant
sound, CT, or MR imaging, however, to help evaluate women [6 – 8].
the renal parenchyma and detect renal masses [4]. Although there continues to be a lack of rigorous
Intravenous urography with nephrotomography can large-scale research on the cost-effectiveness of var-
identify only 21%, 52%, and 82% of masses less than ious urographic imaging strategies in the evaluation
2 cm in diameter, 2 to 3 cm, and 3 cm or larger, of patients with hematuria and other urologic indica-
respectively, when CT is used as the reference stan- tions, sufficient information exists to define a reason-
dard [5]. Moreover, when a mass is detected by able approach to patients with hematuria. This article
intravenous urography, further characterization by describes the evaluation of hematuria and reviews
cross-sectional imaging is necessary because intra- developing concepts and evolving techniques of CT
venous urography cannot reliably distinguish solid and MR urography.
masses from cysts.

Evaluation of patients with hematuria


* Corresponding author.
E-mail address: kawashima.akira@mayo.edu Hematuria is extremely common and can originate
(A. Kawashima). from any site in the urinary tract. The presence of

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00073-3
946 A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961

gross hematuria usually prompts patients to seek niques is whether CT and MR urography can visualize
medical attention, and a thorough urologic investiga- urothelial abnormalities of the intrarenal collecting
tion is warranted to determine its cause. In contrast, systems and ureters with sensitivities equal or superior
the diagnosis, etiology, and management of asymp- to that of conventional intravenous urography.
tomatic microhematuria are controversial. Asymp-
tomatic microscopic hematuria is often not a sign of
underlying surgical urologic disease. Some degree of CT urography
hematuria is identified in 9% to 18% of normal
individuals [9,10]. Routine screening of adults for CT urography is increasingly performed as a de-
microscopic hematuria with dipstick testing is not finitive study for the investigation of hematuria and
recommended because hematuria associated with other urologic indications [13]. The renal parenchyma
significant urologic disease may be intermittent. Once is evaluated with axial CT scans, and then the intra-
asymptomatic microscopic hematuria is documented, renal collecting systems and ureters are visualized by
however, the patients should be evaluated. The defi- one of two general approaches. One approach uses
nition of microscopic hematuria recently recommen- projection radiographs (conventional film-screen ab-
ded by the American Urological Association is three dominal radiographs, computed digital radiographs,
or more red blood cells per high-power field on and CT scanned projection radiographic [SPR]
microscopic evaluation of the urinary sediment from images). A second approach uses thin-section axial
at least two of three properly collected urinalysis CT images obtained during the excretory phase of
specimens [9]. Patients with risk factors for signifi- enhancement with two-dimensional multiplanar refor-
cant urologic disease should be considered for a mation and three-dimensional reconstruction images.
urologic evaluation after one episode of properly These two different CT urographic techniques are both
documented microscopic hematuria [9,11]. These risk attractive because they attempt to combine the sensi-
factors include the development of gross hematuria or tivity and specificity of CT for urinary calculi and
irritable voiding symptoms; a history of smoking or small renal masses with the sensitivity and specificity
chemical exposure; all adults older than 40 years; of intravenous urography for urothelial abnormalities
previous urologic history; a history of urinary tract into one minimally invasive examination [14].
infection or pelvic irradiation; analgesic abuse (eg,
phenacetin); and cyclophosphamide exposure [11]. Combining CT and projection radiography
When hematuria is accompanied by other findings
(eg, proteinuria, red blood cell cast, elevated serum Intravenous urography remains the gold stan-
creatinine), a medical evaluation for the presence of dard for noninvasive visualization of intraluminal
primary diffuse renal parenchymal disease is re- filling defects in the collecting systems and uro-
quired. Patients without these findings and those with thelial abnormalities. Previous series examining
risk factors for significant urologic disease should be patients with hematuria have been based on this
referred promptly for a urologic evaluation [11]. intravenous urography technique. Combined (hy-
Microscopic hematuria associated with anticoagula- brid) CT and conventional intravenous urography
tion therapy frequently is precipitated by significant methods have been implemented to incorporate the
urologic pathology and prompt evaluation is required. strength of each modality into a single examina-
In a study of 1000 consecutive adults with asymp- tion. When projection radiography is used, no CT
tomatic gross or microscopic hematuria by Mariani postprocessing is necessary.
et al [12] in 1989, 9% of patients were found to have In 1996, Perlman et al [15] first described the
life-threatening abnormalities, and an additional 23% concept of CT urography. Conventional intravenous
had lesions requiring at least observation. urograms were obtained in a urography suite, followed
Cystourethroscopy is performed to complete the by patient transfer to a CT suite for supplemental CT
evaluation of the lower urinary tract, primarily the without additional intravenous contrast adminis-
bladder, although gross bladder pathology can be tration, which was limited to the kidneys and any
visualized with imaging studies. Comprehensive up- additional abnormality identified on intravenous urog-
per tract imaging studies should be used to detect raphy (Fig. 1). In 27 of 30 patients with lesions in the
renal cell carcinoma, transitional cell carcinoma, collecting system, the lesions were detected only on
urolithiasis, and renal infection. Because the renal intravenous urography. This technique allowed a rea-
parenchyma is evaluated better by cross-sectional sonable evaluation of the renal parenchyma but lost the
imaging modalities than by intravenous urography, a advantage of unenhanced CT compared with intrave-
primary issue when using these cross-sectional tech- nous urography in detecting urolithiasis.
A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961 947

Fig. 1. Transitional cell carcinoma of the left ureter in a 71-year-old man with a history of gross hematuria. (A) Prone view of
intravenous urogram demonstrates an irregular filling defect in the mid left ureter (arrow) distal to unifying two left moiety ureters
in the partial duplication of the left renal collecting system. (B) CT scan at 3.75-mm slice thickness following the intravenous
urogram (Fig. 1A) without additional intravenous contrast administration reveals a soft tissue mass of the left ureter (arrow)
partially surrounded by contrast material at the level of the aortic bifurcation. (C) Curved planar reformation image demonstrates
the left ureteral mass (arrow). The patient underwent left segmental ureteral resection for noninvasive grade 2 (of 3) papillary
urothelial carcinoma.
948 A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961

In other centers, an abdominal radiograph is first rooms, however, requires additional time, can cause
obtained. Unenhanced and enhanced helical CT scheduling and staffing conflicts, and often affects the
images are next acquired to evaluate renal parenchy- level of pyelocalyceal distention in the urographic
ma and urolithiasis. Finally, the patient is transferred portion of the examination.
to the urography suite to complete the urographic An alternative to this approach, which was imple-
portion of the study. This approach combines the mented at the authors’ center, is the acquisition of
advantages of intravenous urography with those of conventional radiographs with a ceiling-mounted
CT scanning, allowing one comprehensive imaging overhead x-ray tube while the patient is lying on the
study. Movement of the patient between procedure CT table for multiphasic CT acquisitions [16 – 18].

Fig. 2. Transitional cell carcinoma of the right intrarenal collecting system in a 76-year-old woman with gross hematuria.
(A) Intravenous urogram 8 minutes after intravenous contrast material injection with ureteral compression reveals an irregular
round filling defect occupying a mid renal calyx (arrow). (B) Corresponding original CT scanned projection radiographic (SPR)
image obtained at 80 kV and 300 mA demonstrates objectionable dark band along high-density objects, such as iodinated
contrast material. The caliceal mass (arrow). (C) Enhanced CT SPR image after reprocessing the original CT SPR image data
(Fig. 2B) using clinically optimized algorithms substantially minimizes the artifacts and appears similar to that of conventional
intravenous urogram (Fig. 2A). The caliceal mass (arrow). (D) Excretory phase enhanced CT scan at 1.25-mm slice thickness
demonstrates a small soft tissue mass (m) occupying a minor calyx of the interpolar region of the right kidney. (E, F) Coronally
reformatted (E) and thin (8 mm) slab maximal intensity projection (MIP) (F) images demonstrate the relationship of the soft
tissue mass to the intrarenal collecting system. (G) Thick (5 cm) slab average intensity projection (AIP) image demonstrate an
overview of the renal collecting system similar to intravenous urogram (Fig. 2A). The caliceal mass (arrow). The patient
underwent right nephroureterectomy for noninvasive grade 2 (of 3) papillary transitional carcinoma located in a minor calyx.
A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961 949

This method allows high-spatial-resolution intrave- The technique requires the use of an auxiliary, radio-
nous urography films (approximately four line pairs lucent CT tabletop that can accommodate a radio-
per millimeter for film-screen radiography) to be graphic cassette under the patient without introducing
obtained at various times before and after the CT artifacts on the CT image [18]. This approach has been
acquisitions without the need for the patient to move. accepted as an integrated urologic imaging study at

Fig. 2 (continued).
950 A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961

Fig. 2 (continued).

the authors’ institution (Figs. 2, 3) [16]. In the authors’ iodinated contrast media) (see Figs. 2B, 3B) [17].
clinical experience, urothelial abnormalities are better These objectionable artifacts around high-contrast
or only seen on conventional film-screen urograms, objects can be minimized substantially on new im-
which comprise 10% of abnormalities revealed on CT proved CT SPR with reprocessing techniques that use
urography examinations [16]. This underscores the clinically optimized contrast-enhancement algorithms
need for high-quality urographic images in CT urog- while maintaining adequate high-contrast spatial reso-
raphy. Excretory-phase enhanced CT scans can be lution (GE Medical Systems) (see Figs. 2C, 3C)
acquired to correlate with positive or inconclusive [19,20]. This CT urographic approach, combining
projection urographic findings when necessary. helical CT and enhanced CT SPR urographic images,
An alternative method of obtaining projection is attractive because modification of the CT tabletop
images without moving the patient off the CT table and installation of a ceiling-mounted x-ray tube are no
is the use of the CT SPR technique. A CT SPR image, longer necessary, and this technique can be performed
which is referred to as a ‘‘scout view’’ (GE Medical on any multidetector helical CT scanner. As with the
Systems, Milwaukee, WI), ‘‘topogram’’ (Siemens reformatted CT images of the collecting system,
Medical Systems, Iselin, NJ), and ‘‘scanogram,’’ is further scientific studies are needed to validate the
usually used for prescan localization. The spatial sensitivity of optimally enhanced CT SPR images for
resolution of CT SPR (approximately less than one depicting fine urothelial detail.
line pair per millimeter) when obtained at 80 kV and Bowel preparation with a mild laxative before
300mA is inferior to conventional radiography, urography examination usually helps to reduce the
whereas the contrast resolution of opacified structures amount of fecal material and gas in the colon so that
is similar to conventional radiography [17]. Conven- the intrarenal collecting systems can be visualized
tional CT SPR images use simple edge enhancement more clearly with projection radiographs. Diuresis
image filters, however, that result in dark and bright after the ingestion of moderate quantities of fluid or
band artifacts around high-attenuation objects (eg, mild diuretics, such as coffee or tea, decreases the
A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961 951

Fig. 3. Ureteritis cystica in the left ureter in a 60-year-old woman with a history of left renal stone. (A) Intravenous urogram
8 minutes after intravenous contrast injection with abdominal compression demonstrates numerous tiny smooth uniform-sized
filling defects in the left ureter (arrows). (B, C) The appearance of the small urothelial filling defects on enhanced CT SPR image
(C) is more similar to conventional intravenous urogram (Fig. 3A) than original CT SPR image (B) by minimizing the edge
enhancement artifacts. (D) CT scan at 1.25-mm slice thickness obtained during the excretory phase of enhancement demonstrates
a tiny focal intraluminal elevation of the mid left ureteral wall (arrow). (E) Oblique coronal reformatted image demonstrates
many tiny filling defects in the left ureter (arrows). (F, G) The small urothelial filling defects are less well defined on thick (5 cm)
slab MIP (F) and AIP (G) images.

concentration of contrast material in the urinary tract when low-osmolar iodinated contrast material is
during the excretory phase of the examination. This used [1,2]. The intrarenal collecting system and
and further swallowing of gas may be prevented by proximal ureters are well distended on 8-minute
instructing the patient to take nothing by mouth for delayed film, and their appearance with and without
several hours before the examination. compression can be studied [1]. The ureters are
Visualizing the intrarenal collecting system and generally well visualized on 10-minute decom-
ureter with intravenous urography depends on opti- pressed film. Twenty-minute film and postvoid film
mal distention and opacification. After completion are optional and may be useful for the morphologic
of intravenous contrast material injection, ureteral evaluation of the bladder. The evaluation of the
compression is applied unless contraindicated (eg, entire urinary collecting system usually requires
abdominal aortic aneurysm, recent abdominal sur- review of a composite of urographic images.
gery, severe abdominal pain, suspected renal trau- Consideration of radiation exposure is very im-
ma, and urinary diversion or renal transplant) [2]. portant with these new techniques. One abdominal
The use of ureteral compression is important to radiograph with a stationary grid obtained in a 21-cm-
ensure adequate pyelocaliceal distention, especially thick patient delivers an effective skin exposure level
952 A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961

Fig. 3 (continued).

of 412 mR (106.3 mC/kg) and effective dose of SPR image obtained at 300 mA delivered an effective
0.5 mSv [17]. The effective skin exposure and skin exposure level and effective dose of 330 mR
effective doses at conventional radiography must (85.1 mC/kg) and 0.54 mSv [17]. One abdominal-
increase with patient thickness to maintain compara- pelvic CT scan delivers an effective skin exposure
ble image noise, approximately doubling for each level of 2500 mR (645 mC/kg) and effective dose of
additional 4 to 5 cm of patient thickness. One CT approximately 11 mSv. Estimated skin doses and
A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961 953

Fig. 3 (continued).

effective dose for combined CT and projection urog- however, if oral intake is withheld for several hours
raphy are considered to be less than what a separate before the examination.
CT study and conventional intravenous urography In a study using a single-detector helical scanner by
with nephrotomograms deliver. McNicholas et al [21], excretory-phase enhanced CT
scans were obtained with slice thickness of 5 mm, pitch
of 1.5, and slice increment of 2.5 mm. Multidetector
Excretory-phase enhanced CT with multiplanar row helical CT scanners allow single breathhold
reformation and three-dimensional acquisitions of the abdomen and pelvis with narrow
reconstruction imaging collimation to achieve high spatial resolution [27].
Several researchers have used multidetector row CT
This approach relies exclusively on the acquisition with slice thickness of 2.5 to 3 mm and slice increment
of unenhanced and enhanced CT scans of the collect- of 1 to 1.25 mm for CT urography [22,24,25], and
ing systems including the essential acquisition of others more recently used slice thickness increment of
thin-section helical CT scans obtained during the 1 to 1.25 mm to generate a single volume dataset with
excretory phase of enhancement. Multiplanar refor- near-isotropic voxels. Multiplanar reformatted and
mation and three-dimensional reconstruction images three-dimensional reconstructed images can be dis-
obtained during the excretory enhanced phase are played with improved spatial resolution in nontrans-
generated on workstations from axial source images verse planes on workstations [23,26].
[21 – 26]. No bowel preparation is necessary for this Detection of urothelial abnormalities with excre-
type of CT urography examination. The possibility of tory CT urography requires visualization of the
aspiration of solid food by vomiting can be avoided, optimally distended and opacified collecting system
954 A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961

as traditionally seen on intravenous urography. It may of data to be evaluated requires interactive viewing
be difficult to obtain a single set of images on which on a workstation. Multiplanar reformation images
the collecting systems are completely opacified. Sev- provide orthogonal coronal or oblique (en face)
eral studies have shown that CT acquisition using planes, which help to define the location and extent
abdominal compression improved opacification of of the lesions shown on axial CT images (see
the collecting system compared with CT scans without Figs. 2D, 3E). Maximum intensity projection (MIP),
compression [21,28]. Caoili et al [23] used two CT average intensity projection, and perspective volume
acquisitions of the entire urinary tract during the rendering reconstructed images at thin (5 to 20 mm)
excretory phase of enhancement: one with abdominal and thick (35 to 60 mm) slabs can be generated from
compression and the other after release of abdominal the volume data. Thick slab three-dimensional recon-
compression. McNicholas et al [21] showed excretory- structed images provide an overview of the collecting
phase CT scans with patients in a prone position also systems and mimic conventional intravenous uro-
improved opacification of the distal ureters compared grams, but assessment of urothelial wall thickness is
with supine CT scans without abdominal compression. difficult (see Figs. 2G, 3F, 3G). Thin slab recon-
Alternative techniques for achieving optimal visuali- structed images have the advantage of covering a
zation of the collecting systems include supplemental considerably longer range than standard multiplanar
use of normal saline infusion and diuretic injection. reformations and have the ability to demonstrate
McTavish et al [26] reported supplemental infusion small filling defects, which may be obscured by
of 250 mL of physiologic saline immediately after surrounding contrast in the collecting system with
injecting intravenous contrast material significantly thick slab reconstruction (see Fig. 2F). Curved planar
improved opacification of the distal ureters. Nolte- reformation provides a single image to outline the
Ernsting et al [25] reported that intravenous injection course of ureterectasis to the point where an obstruct-
of low-dose diuretics (10 mg of furosemide) before ing process, such as a calculus or tumor, is present
intravenous contrast injection also permitted less (see Fig. 1C) [22]. Large data sets with 400 to 800
dense, homogeneous opacification of the collecting axial source images are common and need to be
systems compared with supplemental infusion of reviewed efficiently at a workstation.
300 mL of normal saline. Because CT has contrast In a study of 65 patients who underwent multi-
resolution superior to conventional radiography, dilu- detector row CT urography for urologic indications,
tion of the contrast material does not substantially CT urography detected 15 of 16 urothelial carcino-
affect perception of contrast enhancement of the mas and many other urinary tract abnormalities [23].
collecting systems [25,26] and may minimize poten- The single missed lesion (transitional cell carcinoma
tial beam-hardening artifacts associated with dense at the bladder base) could be identified in retrospect. In
contrast material in the intrarenal collecting system the same series, three lesions of ureteral transitional
[25]. An alternative CT urographic approach consists cell carcinomas appeared as circumferential ureteral
of two CT acquisitions: unenhanced and enhanced thickening on CT urography. A case of ureteritis had an
[22,24]. Following initial noncontrast CT, two admin- indistinguishable appearance. In a study of 57 patients
istrations of a split dose of intravenous contrast with hematuria and unexplained hydronephrosis who
material are given, and both nephrographic and ex- were studied with both multidetector row CT urogra-
cretory-phase images are acquired during the second phy and retrograde pyelography after nondiagnostic
acquisition [24]. This method, however, requires two intravenous urograms or ultrasonograms, there were
contrast injections and two CT examinations separated 38 intrinsic urothelial lesions including 15 transitional
by 15 minutes. Chow and Sommer [22] reported cell carcinomas, 13 urinary stones, 5 cases of uretero-
obtaining combined nephrographic- and excretory- pelvic junction obstruction, 3 cases of ureteral stric-
phase enhanced CT scans of the kidneys and proximal ture, and 2 bladder polypoid lesions [29]. Of the
ureters with abdominal compression by scanning 90 38 intrinsic lesions, CT urography detected 37 lesions
seconds after the second dose of a two-phase injection with sensitivity of 97%, whereas retrograde pyelo-
of iodinated contrast material temporally separated by grams detected 31 lesions (82%). A bladder tumor was
2 minutes. Immediately after release of compression, missed on CT because the bladder was obscured by
the excretory-phase enhanced CT scan is continued to beam hardening artifacts from bilateral hip prostheses.
cover the distal ureters and bladder. In a study by McTavish et al [26], estimated skin
Assessment of axial CT images (source images), doses from CT urography using the three-phase CT
which are usually displayed with wide window set- scan protocol were similar to standard intravenous
tings similar to the bone window setting, remains urography, whereas the total effective doses from CT
essential for accurate diagnosis, and the large amount urography were approximately two times higher than
A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961 955

intravenous urography. In a study by Caoili et al [23], long T2 relaxation times. Heavily T2-weighted pulse
estimated effective doses from a four-phase CT sequences generate images with high signal intensity
urography protocol ranged 25 to 30 mSv. Estimated from static fluid in the collecting systems, whereas
effective doses from abdominal-pelvic CT and con- the signal intensity from parenchymal tissues with
ventional intravenous urography with nephroto- shorter T2 relaxation times is suppressed. Early
mography were 10 to 15 mSv and 5 to 6 mSv, attempts at MR urography used relaxation enhance-
respectively. The researchers concluded that multi- ment and fast spin echo sequences (GE Medical
phase CT urography exposes a patient to an amount Systems; or turbo spin echo, Siemens Medical Sys-
of radiation similar to what is experienced during a tems, Iselin, NJ, and Philips Medical Systems, Best,
combination of standard intravenous urography and The Netherlands), but these were limited to some
CT of the abdomen and pelvis. Because of the higher extent by the long acquisition times and concomitant
radiation dose, this type of CT urography may be respiratory motion artifacts [6,30]. The most common
indicated for the evaluation of hematuria only in T2-weighted technique currently in use is the single-
patients with a high risk of malignancy. Continued shot fast spin echo sequence (SSFSE, GE Medical
efforts are needed to reduce radiation exposure. Systems; and half-Fourier acquisition single-shot tur-
CT urography takes longer than a standard ab- bo spin echo, Siemens Medical Systems) (Figs. 4 – 6).
dominal-pelvic CT and requires more input and effort This is a variant of fast spin echo in which all of the
from both the technologist and the physician [14]. At 180-degree radiofrequency refocusing pulses needed
the authors’ institution, CT urography (combined CT to generate an image are acquired after a single
and intravenous urography) is charged as a combina- 90-degree excitation. Generally, a half-Fourier acqui-
tion of CT of the abdomen with and without intrave- sition is used so that half of the usual number of phase-
nous contrast, CT of the pelvis with intravenous encoded steps is required [31,32]. This technique
contrast, and a limited intravenous urogram. Three- generates images sequentially within 1 to 2 seconds
dimensional reconstruction is more labor intensive with excellent in-plane spatial resolution. Multiple
and can result in an additional charge. thin-section images can be acquired within a breath-
hold, thereby eliminating respiratory artifact. Thick
section projection images are also useful for global
MR urography evaluation of the urinary tract. Image contrast depends
primarily on the value of TE: as TE is increased,
MR urography can be performed with heavily background suppression increases, but there is a de-
T2-weighted pulse sequences similar to MR cholan- cline in signal-to-noise ratio (SNR).
giopancreatography, T1-weighted pulse sequences Other new pulse sequences may have potential
following intravenous gadolinium contrast agent sim- in MR urography. Steady-state free precession se-
ilar to MR angiography, or a combination of the two. quences (fast imaging using steady-state acquisition
Many studies have documented the ability of MR [FIESTA, GE Medical Systems], fast imaging with
urography to detect urinary tract abnormalities, such steady-state precession [TrueFISP, Siemens Medical
as urinary tract dilatation, ureteric obstruction, dupli- Systems], balanced fast field echo [Philips Medical
cated renal collecting systems, urinary stones, and Systems]) are steady-state coherent gradient echo
urothelial tumors. The sensitivity of renal parenchy- pulse sequences that refocus the transverse magneti-
mal MR imaging with intravenous gadolinium con- zation between the excitation pulse and gradient
trast for assessing renal masses and abnormalities of echo acquisition to minimize substantially dephasing
the nephrogram is considered to be similar to that of effects of transverse magnetization and allow acqui-
CT. Combining renal MR imaging and MR urogra- sition of high SNR images at very short TRs. Steady-
phy can serve as a comprehensive imaging of the state free precession sequences can be performed using
renal parenchyma, collecting systems, and bladder in either two- or three-dimensional acquisitions. Signal
patients who cannot go to routine radiographic stud- intensity of steady-state free precession sequences is
ies, such as pregnant or pediatric patients, patients independent of TR and related to T2/T1, which accen-
with a severe allergy to iodinated contrast media, or tuates signal intensity of fluid. Another sequence is a
patients with impaired renal function. three-dimensional fast recovery fast spin echo (GE
Medical Systems) pulse sequence. The fast recovery
Heavily T2-weighted MR urography feature uses an additional radiofrequency pulse at
the end of each TR to refocus the residual trans-
The fundamental concept underlying this tech- verse magnetization into the z axis, allowing shorter
nique is that simple fluids, such as urine, have very TRs while still enhancing the signal intensity of
956 A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961

Fig. 4. Normal T2-weighted MR urography with and without furosemide. (A) Oblique coronal SSFSE (TR of 2046 milliseconds,
TE of 974 milliseconds, 36  36 cm field of view, matrix of 256  256, 0.5 number of excitation [NEX], 5-cm slice thickness,
2-second acquisition time) demonstrates an extrarenal pelvis ( p) on the right. d = duodenum; s = cerebrospinal fluid; b = bladder.
(B) Repeat SSFSE image 10 minutes after intravenous administration of furosemide (20 mg) reveals distention of the right renal
collecting system ( p), ureter (u), and bladder (b). d= duodenum; s = cerebrospinal fluid; b = bladder.

static fluid and providing high-resolution heavily adequate visualization of the collecting system and
T2-weighted images with shorter acquisition time ureters (see Figs. 4B, 5C) [33 – 35].
compared with three-dimensional fast spin echo Heavily T2-weighted MR urograms can be
imaging. In general, three-dimensional sequences obtained as single thick (5 to 9 cm) slab projection
offer fundamental advantages over two-dimensional images or MIP views generated from multiple thin
acquisitions: SNR is usually higher, through-plane (5 mm or less) section images. Single thick slab
spatial resolution is improved, and there is less oppor- projection imaging with a large field of view obtained
tunity for spatial misregistration artifacts caused by in coronal and both oblique coronal projections
breathholding discrepancies. provides a quick survey of the upper tract without
These heavily T2-weighted MR urography tech- requiring any postprocessing. The multislice method
niques allow for excellent visualization of the urinary is more time-consuming, but the acquisition of thin
tract and are particularly useful in patients with single slices reduces partial-volume averaging and
dilated collecting systems. T2-weighted MR urogra- offers a better opportunity to detect small intraluminal
phy is suitable for the detection and localization of filling defects, which may be obscured by surround-
ureterectasis, especially in the case of markedly ing urine, particularly in the dilated system on a
impaired renal function, where contrast-enhanced single thick slab projection MR urogram.
urography is limited by impaired excretion of con- Visualization of the urinary tract may be degraded
trast. If distention of the collecting system is inade- by superimposition of fluid-filled extraurinary struc-
quate, it is difficult completely to visualize anatomic tures (eg, bowel loops, common bile duct, gallblad-
detail. Adequate hydration is essential for diagnostic der); this is most problematic with thick-slice heavily
MR urography. In many patients, particularly with T2-weighted MR urograms. Thin-slice images are not
nonobstructed, nondilated collecting systems, the affected by superimposed extraurinary fluid-filled
addition of diuretics (eg, furosemide) is critical for structures. The image quality of three-dimensional
A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961 957

Fig. 5. Normal T2-weighted MR urography and gadolinium-enhanced T1-weighted MR urography. (A) Intravenous urogram
demonstrates normal renal collecting systems. (B, C) Heavily T2-weighted SSFSE image (TR of 2046 milliseconds, TE of
974 milliseconds, 5-cm slice thickness, 36  36 cm field of view, NEX of 0.5, matrix of 256  256, 2-second acquisition time)
after intravenous injection of furosemide (C) demonstrates improved distention of the intrarenal collecting system when
compared with heavily T2-weighted SSFSE image before furosemide injection (B). (D) Gadolinium-enhanced three-dimensional
SPGR (TR of 6.4 ms, TE of 1.4 ms, flip angle of 45 degrees, 26  19.5 cm field of view, 1.6-mm slice thickness, 0.8-mm
overlap, 33-s acquisition time, 256  160  1 NEX) demonstrates improved visualization of the renal collecting systems.

MIP reconstructions can be improved by manually tract (GE Medical Systems; fast low-angle shot,
removing extraurinary fluid-containing structures Siemens Medical Systems; fast-field echo, Philips
from the volume. Medical Systems) [34,38] is an alternative method
A significant limitation of heavily T2-weighted of MR urography and is also referred to as ‘‘contrast-
MR urography is that it provides relatively little enhanced excretory MR urography’’ (see Fig. 5D;
functional information [36]. It can be difficult at Fig. 7). Three-dimensional sequences generate low
times to distinguish between obstructive and non- contrast images of parenchymal and background tis-
obstructive urinary dilatation. One clue often noted in sue, an effect accentuated as the flip angle increases.
cases of acute obstruction is the presence of perirenal Image contrast is provided by the T1-shortening
edema, seen as high signal intensity on T2-weighted properties of gadolinium as it is excreted into the
images [37]. urinary collecting systems. Additional background
suppression can be provided by the use of spectral
Gadolinium-enhanced T1-weighted MR urography fat saturation. An alternative, little used technique is to
obtain both precontrast and postcontrast three-dimen-
Gadolinium-enhanced T1-weighted three-dimen- sional acquisitions and then subtract the mask from
sional spoiled gradient echo imaging of the urinary the postcontrast data set. This technique provides
958 A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961

Fig. 6. Papillary necrosis in a 45-year-old woman with sickle cell trait, who presented with asymptomatic microhematuria.
(A) Intravenous urogram obtained 10 minutes after intravenous contrast administration demonstrates multiple areas of contrast
pooling in the papillary regions of the left kidney adjacent to calyces (arrows). (B) Heavily T2-weighted SSFSE (TR of
2046 milliseconds, TE of 887 milliseconds, field of view of 16  26 cm, matrix of 256  256, NEX of 0.5, 5-cm slice thick,
2-second acquisition time) using torso phase array coil 10 minutes following furosemide injection with ureteral compression
demonstrates paracaliceal papillary cavities (arrows).

excellent background suppression but is limited by the enhanced T1-weighted MR urograms can provide
potential for misregistration artifacts. Acquisition of gross assessment of renal function. Suboptimal opa-
contrast-enhanced MR urography during a breathhold cification of the urinary tract (eg, markedly impaired
is extremely important to eliminate motion artifact renal function and high-grade urinary obstruction) can
caused by respiratory excursion of the kidneys limit the value of this technique for morphologic
[38,39]. Respiratory-gated imaging is an alternative assessment. MIP images are generated from excreto-
when respiratory suspension is difficult (eg, pediatric ry-phase enhanced three-dimensional spoiled gradient
patients) [40,41]. Images are typically acquired images. Source images are essential to detect subtle
5 to 8 minutes after intravenous administration of abnormalities of the urinary tract, such as small intra-
gadolinium contrast agent. Contrast-enhanced three- luminal filling defects that may be obscured by
dimensional MR urography can be improved by using opacified urine on MIP images.
a diuretic (eg, furosemide) to provide additional A major limitation of MR urography is the detec-
distention and improved visualization of the collecting tion of urinary calculi, which generally appear as
system and ureters (see Fig. 5D) [42]. Diuretic is filling defects or signal voids on both heavily T2-
useful not only to distend the collecting system but weighted and contrast-enhanced three-dimensional
also optimally to dilute the concentration of gado- spoiled gradient images. Jung et al [43] investigated
linium in the urinary tract, which helps avoid signal 82 patients with ureteric obstruction shown or sus-
loss caused by T2* effects associated with concen- pected at intravenous urography with heavily T2-
trated gadolinium in urine. With thick (35 to 50 mm) weighted and gadolinium-enhanced T1-weighted MR
section T1-weighted two-dimensional spoiled gra- urograms. Of 72 patients with ureteral stones, intra-
dient sequences, contrast-enhanced MR urography venous urography and MR urography correctly di-
provides an overview of the collecting system includ- agnosed 49 and 64, respectively. Of eight patients
ing the intrarenal collecting system [38]. Gadolinium- with ureteric tumors, intravenous urography and MR
A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961 959

sign in predicting acute ureteric obstruction. The MR


imaging findings of a nonenhancing filling defect in
the ureter are not specific for a ureteral stone,
however, and may represent blood clot, gas, fungus
ball, and sloughed papilla. Furthermore, it is quite
difficult to visualize small nonobstructive ureteral
calculi and small caliceal renal calculi at MR urog-
raphy with current techniques. On occasion, the
distinction of ureteral stone from vascular impression
and physiologic peristalsis of the ureter may be
problematic. Because urinary stones are not directly
visualized at MR urography, MR urography can be
combined with unenhanced CT to assess for calculi
in patients with hematuria.
Another limitation of MR urography is its rela-
tively poor spatial resolution [36]. Current MR urog-
raphy techniques do not provide visualization of
anatomic detail of the calyces, infundibula, and
ureters equivalent to intravenous urography or CT
urography. It is possible that subtle urothelial abnor-
malities, such as small malignancies, may be unde-
tected with MR urography.
MR urography is, however, an evolving technique
that offers great promise in its ability to provide both
anatomic and functional information. The tradeoffs
between acquisition time, SNR, and spatial resolution
will be alleviated to some extent by the introduction
of parallel imaging techniques to increase acquisition
Fig. 7. Hydronephrosis in a renal transplant. Gadolinium- speed, improved phased-array coil design, new and
enhanced three-dimensional SPGR MIP image demonstrates refined pulse sequences, and the availability of high-
pyelocaliectasis and ureterectasis of the transplanted kidney field (3 T) imaging systems to increase SNR. The
with a short segment of kinking of the distal ureter (arrow) unique ability of MR imaging to provide quantitative
at the insertion into the native bladder (b). functional information, such as blood flow, perfusion,
and glomerular filtration rate, in addition to anatomic
urography correctly diagnosed three and seven, re- characterization of the parenchyma and collecting
spectively. One of the ureteric tumors was misdiag- system could lead to a single comprehensive diag-
nosed as a stone by MR urography, because there nostic study [41,44].
was no appreciable contrast enhancement identified.
The main reason for the failure of intravenous
urography was most often the absence of contrast
medium excretion. Dilatation of the urinary tract Summary
facilitated visualization of an intrinsic or extrinsic
obstructing lesion with MR urography. In a recent CT urography and MR urography are an evolving
study of 49 patients with acute flank pain who were concept and developing technique. As the technology
studied by unenhanced CT, MR urography, and matures, CT urography will combine the ultimate
intravenous urography, ureteral stones were present diagnostic capabilities of intravenous urography and
in 32 patients [39]. When a complete or partial filling CT. In the near future, many intravenous urograms
defect within the urinary tract on both heavily T2- will be replaced by CT urography to evaluate patients
weighted and gadolinium-enhanced T1-weighted MR with hematuria and other genitourinary conditions.
urographic sequences was presumed to be a stone, MR urography currently serves as an alternative
the sensitivity and specificity of MR urography was imaging technique to intravenous urography and
94% to 100%, and 100% in diagnosing ureteral CT urography for children and pregnant women
stones. Perirenal edema shown as high signal inten- and for patients with contraindications to iodinated
sity on T2-weighted images was a useful secondary contrast media.
960 A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961

Acknowledgments [14] Herts BR. The current status of CT urography (2002).


Crit Rev Comput Tomogr 2002;43:219 – 41.
The authors thank Andrew J. LeRoy, MD, and [15] Perlman ES, Rosenfield AT, Wexler JS, Glickman MG.
CT urography in the evaluation of urinary tract disease.
Robert R. Hattery, MD, for their editorial assistance
J Comput Assist Tomogr 1996;20:620 – 6.
and Kathryn A. Herman for assistance with manu-
[16] Vrtiska TJ, King BF, LeRoy AJ, Hattery RR, McCol-
script preparation. lough CH, Quam JP. CT urography: analysis of tech-
niques and comparison with IVU [abstract]. Radiology
2000;217:225.
References [17] McCollough CH, Bruesewitz MR, Vrtiska TJ, et al.
Image quality and dose comparison among screen-
[1] Hattery RR, Williamson Jr B, Hartman GW, LeRoy AJ, film, computed, and CT scanned projection radiogra-
Witten DM. Intravenous urographic technique. Radiol- phy: applications to CT urography. Radiology 2001;
ogy 1988;167:593 – 9. 221:395 – 403.
[2] Dyer RB, Chen MY, Zagoria RJ. Intravenous urogra- [18] McCollough CH, Daly TR, King Jr BF, LeRoy AJ. An
phy: technique and interpretation. Radiographics 2001; auxiliary CT tabletop for radiography at the time of
21:799 – 821. CT. J Comput Assist Tomogr 2001;25:876 – 80.
[3] Smith RC, Rosenfield AT, Choe KA, et al. Acute flank [19] McCollough CH, Hsieh J, Vrtiska TJ, King BF, LeRoy
pain: comparison of non-contrast-enhanced CT and in- AJ, Fox SH. Development of an enhanced CT digital
travenous urography. Radiology 1995;194:789 – 94. projection radiograph for uroradiologic imaging. Radi-
[4] Amis Jr ES. Epitaph for the urogram. Radiology 1999; ology 2001;218:609.
213:639 – 40. [20] Kawashima A, Vrtiska TJ, King BF, et al. Improved
[5] Warshauer DM, McCarthy SM, Street L, et al. Detec- CT scanned projection radiographs (SPRs) utilizing
tion of renal masses: sensitivities and specificities of enhanced algorithms: can improved CT SPRs replace
excretory urography/linear tomography US, and CT. conventional film-screen radiographs for CT urography
Radiology 1988;169:363 – 5. [abstract]? Radiology 2001;221:501.
[6] Sigmund G, Stoever B, Zimmerhackl LB, et al. RARE- [21] McNicholas MM, Raptopoulos VD, Schwartz RK, et al.
MR-urography in the diagnosis of upper urinary tract Excretory phase CT urography for opacification of the
abnormalities in children. Pediatr Radiol 1991;21: urinary collecting system. AJR Am J Roentgenol 1998;
416 – 20. 170:1261 – 7.
[7] Roy C, Saussine C, Jahn C, et al. Fast imaging MR [22] Chow LC, Sommer FG. Multidetector CT urography
assessment of ureterohydronephrosis during pregnancy. with abdominal compression and three-dimensional re-
Magn Reson Imaging 1995;13:767 – 72. construction. AJR Am J Roentgenol 2001;177:849 – 55.
[8] Klein LT, Frager D, Subramanium A, Lowe FC. Use [23] Caoili EM, Cohan RH, Korobkin M, et al. Urinary
of magnetic resonance urography. Urology 1998;52: tract abnormalities: initial experience with multi-detec-
602 – 8. tor row CT urography. Radiology 2002;222:353 – 60.
[9] Grossfeld GD, Litwin MS, Wolf JS, et al. Evaluation [24] Chai RY, Jhaveri K, Saini S, Hahn PF, Nichols S,
of asymptomatic microscopic hematuria in adults: the Mueller PR. Comprehensive evaluation of patients
American Urological Association best practice policy – with haematuria on multi-slice computed tomography
part I: definition, detection, prevalence, and etiology. scanner: protocol design and preliminary observations.
Urology 2001;57:599 – 603. Australas Radiol 2001;45:536 – 8.
[10] Mohr DN, Offord KP, Owen RA, Melton III LJ. [25] Nolte-Ernsting CC, Wildberger JE, Borchers H,
Asymptomatic microhematuria and urologic disease: Schmitz-Rode T, Gunther RW. Multi-slice CT urogra-
a population-based study. JAMA 1986;256:224 – 9. phy after diuretic injection: initial results. Rofo Fortschr
[11] Grossfeld GD, Litwin MS, Wolf Jr JS, et al. Evaluation Geb Rontgenstr Neuen Bildgeb Verfahr 2001;173:
of asymptomatic microscopic hematuria in adults: the 176 – 80.
American Urological Association best practice policy – [26] McTavish JD, Jinzaki M, Zou KH, Nawfel RD, Silver-
part II: patient evaluation, cytology, voided markers, man SG. Multi-detector row CT urography: compari-
imaging, cystoscopy, nephrology evaluation, and fol- son of strategies for depicting the normal urinary
low-up. Urology 2001;57:604 – 10. collecting system. Radiology 2002;225:783 – 90.
[12] Mariani AJ, Mariani MC, Macchioni C, Stams UK, [27] Hu H, He HD, Foley WD, Fox SH. Four multidetector-
Hariharan A, Moriera A. The significance of adult row helical CT, image quality and volume coverage
hematuria: 1,000 hematuria evaluations including a speed. Radiology 2000;215:55 – 62.
risk-benefit and cost-effectiveness analysis. J Urol [28] Caoili EM, Cohan RH, Korobkin M, et al. Effective-
1989;141:350 – 5. ness of abdominal compression during helical renal
[13] Newhouse JH, Bluth EI, Bush Jr WH, et al. Radio- CT. Acad Radiol 2001;8:1100 – 6.
logical investigation of patients with hematuria. In: [29] McCarthy CL, Cowan NC. Multidetector CT urography
ACR Appropriateness Criteria. Reston (VA): American (MD-CTU) for urothelial imaging [abstract]. Radiology
College of Radiology; 2001. p. 1 – 5. 2002;225:137.
A. Kawashima et al / Radiol Clin N Am 41 (2003) 945–961 961

[30] Friedburg HG, Hennig J, Frankenschmidt A. RARE- comparison of conventional gradient-echo sequences
MR urography: a fast nontomographic imaging proce- and echo-planar imaging. Eur Radiol 2001;11:
dure for demonstrating the efferent urinary pathways 18 – 27.
using nuclear magnetic resonance. Radiologe 1987;27: [39] Sudah M, Vanninen R, Partanen K, Heino A, Vainio P,
45 – 7. Ala-Opas M. MR urography in evaluation of acute flank
[31] Regan F, Bohlman ME, Khazan R, Rodriguez R, pain: T2-weighted sequences and gadolinium-enhanced
Schultze-Haakh H. MR urography using HASTE three-dimensional FLASH compared with urography.
imaging in the assessment of ureteric obstruction. Fast low-angle shot. AJR Am J Roentgenol 2001;176:
AJR Am J Roentgenol 1996;167:1115 – 20. 105 – 12.
[32] Tang Y, Yamashita Y, Namimoto T, et al. The value of [40] Staatz G, Rohrmann D, Nolte-Ernsting CC, et al. Mag-
MR urography that uses HASTE sequences to reveal netic resonance urography in children: evaluation of
urinary tract disorders. AJR Am J Roentgenol 1996; suspected ureteral ectopia in duplex systems. J Urol
167:1497 – 502. 2001;166:2346 – 50.
[33] Rothpearl A, Frager D, Subramanian A, et al. MR uro- [41] Rohrschneider WK, Haufe S, Wiesel M, et al. Func-
graphy: technique and application. Radiology 1995; tional and morphologic evaluation of congenital urinary
194:125 – 30. tract dilatation by using combined static-dynamic MR
[34] Nolte-Ernsting CC, Bucker A, Adam GB, et al. Gado- urography: findings in kidneys with a single collecting
linium-enhanced excretory MR urography after low- system. Radiology 2002;224:683 – 94.
dose diuretic injection: comparison with conventional [42] King BF, Der DH, Hattery RR, LeRoy AJ, Williamson
excretory urography. Radiology 1998;209:147 – 57. Jr B, Riederer SJ. Breathhold 3D-SPGR MR urography
[35] Nolte-Ernsting CC, Adam GB, Gunther RW. MR utilizing furosemide and gadolinium: a new MR urog-
urography: examination techniques and clinical appli- raphy [abstract]. Radiology 2000;217:224.
cations. Eur Radiol 2001;11:355 – 72. [43] Jung P, Brauers A, Nolte-Ernsting CA, Jakse G,
[36] Hattery RR, King BF. Technique and application of Gunther RW. Magnetic resonance urography enhanced
MR urography. Radiology 1995;194:25 – 7. by gadolinium and diuretics: a comparison with con-
[37] Regan F, Petronis J, Bohlman M, Rodriguez R, Moore ventional urography in diagnosing the cause of ureteric
R. Perirenal MR high signal – a new and sensitive obstruction. BJU Int 2000;86:960 – 5.
indicator of acute ureteric obstruction. Clin Radiol [44] Lee VS, Rusinek H, Johnson G, Rofsky NM, Krinsky
1997;52:445 – 50. GA, Weinreb JC. MR renography with low-dose gado-
[38] Nolte-Ernsting CC, Tacke J, Adam GB, et al. Diu- pentetate dimeglumine: feasibility. Radiology 2001;
retic-enhanced gadolinium excretory MR urography: 221:371 – 9.
Radiol Clin N Am 41 (2003) 963 – 978

Renal imaging with ultrasound contrast: current status


Michelle L. Robbin, MDa,*, Mark E. Lockhart, MD, MPHa,
Richard G. Barr, PhD, MDb
a
Department of Radiology, University of Alabama at Birmingham, 619 19th Street South, JTN363,
Birmingham, AL 35249 – 6830, USA
b
Department of Radiology, St. Elizabeth’s Health Center, 1044 Belmont Avenue, Youngstown, OH 44501 – 1790, USA

The use of ultrasound contrast agents (UCAs) in saline for echocardiography of the aortic root [3].
the kidney has great promise, but has been less com- Further research led to intravenously injected micro-
pletely studied than in the heart and liver [1,2]. In part, bubbles that were generally large and were trapped by
this is because there is less ambiguity regarding the the pulmonary capillary bed. Early contrast attempts
cause and treatment of renal masses as compared with thus yielded substantial enhancement in only the right
liver masses. Another major reason why renal contrast atrium and ventricle. The room air microbubbles
ultrasound (US) has lagged heart and liver applications created were short lived because they lacked a shell.
is that multiple competing modalities exist that per- A shell provides both stability and a relative barrier to
form well in such areas as renal artery stenosis and limit the diffusion of the gas within the microbubble
renal trauma. There are several clinical scenarios, into the surrounding plasma.
however, in which renal US contrast imaging may The next innovations were to control the size of
become the low-cost noninvasive modality of choice: the microbubble to less than 7 mm to traverse the
the indeterminate renal lesion, complex cyst evalua- pulmonary circulation, and to use an inert gas for the
tion, and the evaluation of pyelonephritis. microbubble rather than room air. The use of an inert
The status of the UCAs currently available for gas not readily soluble in blood plasma increased the
clinical and investigative use is reviewed. A brief in- longevity of the microbubble, called persistence. The
troduction to the microbubble-specific US techniques addition of various microbubble shell compositions to
useful in renal evaluation is given. Current literature coat and further stabilize the microbubble achieved
and experience with the evaluation of a large number persistence in the peripheral circulation on the order
of renal abnormalities are then presented in a problem- of several minutes or more [4].
based format. Clinically useful microbubbles must be strong
enough to withstand passage through the lungs,
capillaries, and exposure to the pressures generated
US contrast agents in the left ventricle. The ideal UCA should also be
injectable intravenously, easy to use, and without
Current UCAs consist of intravenously injected significant side effects. In particular, it should not
microbubbles. These microbubbles substantially in- be nephrotoxic, so as to be a substitute contrast agent
crease the number of reflectors in the vascular space, for patients with elevated creatinine and those allergic
dramatically increasing backscatter in vessels. The to iodine-based CT contrast agents. In general, these
first microbubbles used clinically were created from UCAs remain within the intravascular space in the
kidney, and are not excreted into the collecting
system, in contradistinction to CT contrast agents.
Although CT is extremely useful in radiologic
* Corresponding author. imaging overall, few modalities can compete with
E-mail address: mrobbin@uabmc.edu (M.L. Robbin). US in real-time imaging of the heart. The development

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00070-8
964 M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978

Table 1
Summary of UCA approval by country
Contrast agent Pharmaceutical Location Indications Countries approved
Imagent Alliance Pharmaceutical San Diego, CA Echocardiography United States
SonoVue Bracco Diagnostics Princeton, NJ Echocardiography Europe
Definity Bristol-Myers Squibb North Billerica, MA Echocardiography, United States, Canada
echocardiography of
liver and kidney
Optison Amersham Health Princeton, NJ Echocardiography United States, Europe, Canada
Levovist Schering Berlin, Germany Echocardiography, More than 40 countries in Europe,
vascular imaging Asia (including Japan), Latin
America, Canada, and Australia

of UCAs was initially aimed at cardiac imaging. US, contrast agents could not be detected in the
Table 1 lists UCAs that have been approved in at least microcirculation of organs, such as the liver and
one country or geographic area as of this writing kidney [8].
(March 2003). Optison, Definity, and Imagent are An imaging revolution occurred with the appli-
the three Food and Drug Administration (FDA) – cation of harmonic imaging techniques to contrast
approved UCAs in use in the United States. These agents [9]. Harmonic imaging is performed in gray
agents are approved for use in patients with suboptimal scale instead of color Doppler, with the advantages of
echocardiograms, to opacify the left ventricular cham- the increased spatial and temporal resolution seen in
ber, and to improve the delineation of the left ventricu- gray-scale imaging. Harmonic imaging takes advan-
lar endocardial border. tage of the fact that microbubbles vibrate when
There are currently no FDA-approved UCAs for insonated with the alternating positive and negative
noncardiac applications in the United States. Many pressures of the sinusoidal sound waves [8]. If ex-
US laboratories have been performing evaluations of posed to sufficient energy in the incoming US signal,
the kidney and liver in clinical trials in multiple microbubbles exhibit nonlinear behavior, because
countries. The authors and others have been success- they can expand much more than they can contract.
fully applying this technology to solve relevant Fortunately, the resonant frequency of microbubbles
clinical problems primarily in the liver and kidney, small enough to traverse the pulmonary and capillary
using off-label UCAs that are FDA-approved for circulations (< 7 mm) falls within the diagnostic range
cardiac imaging. Widespread acceptance and use of of US, from 1 to 10 MHz [10]. The harmonic portions
UCAs in the United States is not likely to occur until of the image are further separated from the fundamen-
FDA approval for radiologic indications is achieved, tal (nonharmonic) information by various techniques
and adequate drug reimbursement by insurance com- that involve US phase or pulse manipulation.
panies occurs. Multiple pulses that vary in either pulse direction
or phase are sent down the same or adjacent scan
lines, depending on the manufacturer. An example of
Microbubble-specific imaging techniques one method is as follows: a pulse is sent down a
single scan line. Next, the exact opposite pulse is sent
Early on, the use of UCAs tended to focus on the down the same scan line. Returning echoes from the
microbubble as a rescue agent, to be used only in an insonated tissues, which behave linearly, cancel out
otherwise failed US examination. These initial appli- when added at the scanner. Returning echoes from the
cations were primarily performed with color and nonlinear microbubble interactions do not cancel
power Doppler. Post-US contrast evaluation con- when summed, and give a signal related primarily
sisted of analysis of discrete vessels, determining if to the harmonic signature of the microbubble. This
US contrast increased one’s ability to detect smaller process results in a substantial increase in the signal-
or deeper vessels in a renal or liver lesion [5] or to-noise ratio of the blood with respect to surrounding
increase the success rate of vessel visualization, such tissue [10].
as the renal artery [6,7]. Although impressive results Interaction of the US beam and microbubble is
were obtained in visualizing smaller vessels and dependant on the microbubble gas and shell composi-
deeper macrovasculature with respect to conventional tion, in that a fragile bubble shell may take less power
M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978 965

to break. This is a complex problem, because bubbles It can be argued that large solid masses that are
that are too stiff may be broken more easily traversing isoechoic or hypoechoic to renal parenchyma do not
the pulmonary circulation or capillaries. In general, a need further evaluation with any contrast media.
low-power (low mechanical index [MI]) technique Invariably, however, these patients have a CT scan
creates a harmonic signature that can be imaged before surgery, to address several important considera-
readily, but has a low incidence of bubble breakage. tions: (1) the presence or absence of tumor extension
Low MI imaging can be used to view arterial into the renal vein, important for surgical planning; (2)
inflow into an organ, such as the kidney. Relative evaluation of the contralateral kidney for synchronous
vascularity can be assessed by the echogenicity of renal tumor, which occurs in approximately 2% of
tumor versus normal adjacent parenchyma. High MI patients [20]; (3) staging of the rest of the abdomen,
imaging at higher power insonation (still within FDA assessing for metastases, particularly within the liver.
limits) disrupts the microbubbles, and is another All of these reasons for imaging the patient with
useful technique in US contrast imaging. Once all CT before surgery can be accomplished with con-
the microbubbles are broken in a given portion of trast-aided US. A US contrast study may be useful to
tissue, additional microbubbles are allowed to wash increase diagnostic confidence in the diagnosis of
in over a given time interval. Another high MI pulse renal cell carcinoma in the incidentally detected solid
is then performed. This technique allows an assess- renal mass, before any other imaging tests are per-
ment of relative parenchymal vascularity over a given formed, and before the patient leaves the US labora-
time interval on still images. Such interval delay tory. Further study is necessary, however, to compare
images obviate the necessity for the formal compu- the accuracy, sensitivity, and specificity of renal mass
tation of wash in or washout curves, or region-of- characterization with US contrast with CT.
interest comparisons.
Renal cell carcinoma: detection
It is a relatively common scenario to have searched
Uses of renal US contrast
a kidney for tumor at US, only to have a small lesion
found on a subsequent CT. This is because US is less
Renal mass evaluation
sensitive in detecting small renal lesions, particularly if
they are noncontour deforming [21]. Jamis-Dow et al
Renal cell carcinoma: evaluation
[22] found that CT depicts more renal masses and
The most common application of UCAs in the
smaller renal masses than US. Of lesions that were less
kidney is in renal mass detection and evaluation. Renal
than 1 cm, 24% were missed by CT versus 80% missed
tumors represent 2.6% of newly diagnosed malignan-
at US. For a lesion size of 2 cm, 5% were missed with
cies [11]. Although approximately 2% of autopsies
CT and 30% missed at US. When lesions reached a
may detect renal cell carcinoma [12], it is uncertain as
diameter of 3 cm, 99% were detected with CT and 95%
to how many represent clinically significant disease.
with US.
The survival for surgically treated stage 1 disease (con-
Detection of small renal masses with US contrast is
fined to the kidney) is excellent. When greater than
aided by observing an alteration of the normal cortical
5 cm diameter, however, only 36% of renal cell car-
thickness and renal pyramid spacing by a mass. The
cinomas are stage 1, as compared with 63% of smaller
renal mass depicted in Fig. 1 was not seen on conven-
tumors [13]. The previous 5-year survival rate of stage
tional US. Post-US contrast, an obvious large mass
1 renal cell carcinoma has been 67% to 73% [14,15].
was seen, altering the normal spacing of the renal
More contemporary survival rates using revised TNM
pyramids and cortex, and extending into the renal
staging have been 91%, 74%, 67%, and 32% for stage
hilum. US contrast may help to substantially improve
I, II, III, and IV disease, respectively [16].
ultrasound’s ability to detect small renal masses, par-
Benign lesions, such as oncocytoma, may appear
ticularly those that are noncontour deforming. It is
similar to renal cell carcinoma at imaging. Renal cell
uncertain whether US with contrast will be as good in
carcinomas, however, make up 90% to 95% of malig-
the detection of the small renal cell carcinoma as CT or
nant renal neoplasms, and oncocytomas constitute
MR imaging. It will, however, likely narrow the gap
only about 3% of renal neoplasms [17]. Because there
reported previously.
is no way reliably to distinguish these two entities,
these tumors need to be removed surgically when pos-
sible [18]. Renal cell carcinomas may be less vascular, Angiomyolipoma
hypervascular, or similar in vascularity to adjacent The most common cause for an echogenic renal
cortex when imaged with UCAs, as on CT [19]. mass is an angiomyolipoma (AML). As the spatial
966 M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978

Fig. 1. A 40-year-old woman for evaluation of incidental renal lesion on CT. (A) Conventional longitudinal gray-scale ultrasound
image of the right kidney has a normal appearance. (B) Postcontrast high mechanical index (MI) image of the same region shows
a large enhancing central renal mass (arrows). (C) Longitudinal postcontrast low MI image demonstrates the normal pyramid
distribution (arrows), which is disrupted by a vascular renal mass (arrowheads)

resolution of US, CT, and MR imaging improves, very echogenic renal cell carcinoma with the AML,
however, smaller renal tumors are being detected however, ensures that CT will continue to be per-
more frequently. These tumors are often incidentally formed in all echogenic renal masses. Thin-section
found during the growing numbers of examinations noncontrast CT can identify the presence of intra-
performed for a nonurologic indication [21]. Forman tumoral fat, characteristic of AMLs, which is rarely
et al [23] report that 77% of small renal tumors (< 3 seen in renal cell carcinomas [26].
cm) were either slightly or markedly hyperechoic, A recent investigation using Levovist did not find
compared with 32% of the larger tumors. any improvement in diagnostic accuracy in contrast-
Demonstration of an anechoic rim and intratu- aided characterization of the hyperechoic renal mass
moral cysts can aid in the differentiation of renal cell over that of conventional US [27]. Power Doppler
carcinoma from AML [24,25]. Yamashita et al [24] was used to characterize vascular patterns, however,
hypothesized that the anechoic rim was compressed rather than gray-scale contrast imaging. It is interest-
surrounding renal tissue. The anechoic areas with ing to note that a combined noncontrast and contrast
through sound transmission were thought to be cystic evaluation similar to that performed in CT and MR
changes in the tumor. Intratumoral cysts and anechoic imaging is necessary in the evaluation for hyper-
rims were only in renal cell carcinomas, and not in echoic renal tumors. Fig. 2 shows a patient with
the AMLs. The substantial overlap of the mildly or multiple AMLs. On gray-scale imaging postcontrast,
M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978 967

Fig. 2. A 54-year-old woman with diabetes and incidental echogenic renal lesions on ultrasound. (A) Conventional oblique gray-
scale image of the kidney demonstrates multiple echogenic renal angiomyolipomas (arrows). (B) Oblique postcontrast ultrasound
using low mechanical index does not demonstrate the lesions because of increased echogenicity of the surrounding renal
parenchyma. There is focal alteration of the renal contour (arrows), which represents an angiomyolipoma.

these lesions blend in with the adjacent vascular Cystic masses


cortex, effectively obscuring the lesions. Renal cysts are common incidental lesions found
It is possible that the hypervascular nature of at US, CT, and MR imaging performed for other
the typical renal cell carcinoma may allow differ- indications. Simple cysts are benign and have no
entiation from an AML using gray-scale imaging. malignant potential. Complex cysts vary in their
Fig. 3 demonstrates the typical hypervascular pat- malignant potential, depending on the number and
tern of a renal cell carcinoma that is moderately thickness of septations, the presence of mural nod-
hyperechoic to adjacent renal parenchyma on con- ules, and peripheral calcification. The degree of
ventional US. contrast enhancement of the septations and mural

Fig. 3. A 37-year-old woman with incidental echogenic renal lesion on conventional ultrasound for epigastric pain.
(A) Conventional longitudinal gray-scale image of the kidney shows a moderately echogenic renal mass (arrows) extending from
the medullary region to the cortex. (B) Postcontrast high mechanical index image of the same region demonstrates enhancement of
the lesion (arrowheads), which is hypervascular relative to the adjacent parenchyma. Pathology confirmed renal cell carcinoma.
968 M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978

nodules is a key factor in determining whether the important to realize, however, that some minimal
lesion needs to be removed surgically or merely septation enhancement may occur. Undoubtedly, a
followed [28]. It can be difficult to demonstrate small amount of septation vascularity is present or
vessels in a typical solid renal cell carcinoma at the septation necroses and degenerates. A few small
US, despite low-flow settings on current equipment bubbles of contrast traveling in a septation are unlikely
and the demonstration of substantial vascularity at CT to represent a malignancy, in the authors’ opinion.
or MR imaging. Similarly, it is extremely unusual to Even with thin CT slices through a renal tumor, partial
detect vessels in mural nodules or septations in a volume average can occur, sufficient to mask relatively
complex cyst on conventional US, unless the lesion is thin but enhancing septations. Significant enhance-
obviously a cystic renal cell carcinoma. ment of a cyst septation with microbubbles is always
Ultrasound contrast has been shown to improve worrisome for tumor, even if the CT is negative or
the sonographic depiction of vascularity within intra- indeterminate. Significant septation or mural nodule
cystic septations or solid components, using Levovist enhancement, however, is not 100% specific for ma-
and power Doppler [29]. In this small study of lignancy [30].
13 patients, if power Doppler signals were found in Finer intracyst detail is commonly seen at US.
any part of the cyst either precontrast or postcontrast Cyst contents typically look more complex at US
US, the lesion was considered to be malignant. Six than on CT, altering the renal cyst morphologic
out of the 12 patients who showed no power Doppler classification somewhat from the Bosniak classifica-
signals on conventional power Doppler had markedly tion used with CT. A suggested classification and
enhanced power Doppler signals after contrast injec- work-up scheme for a renal cystic lesion using UCAs
tion. One additional patient was classified as a malig- is as follows [1], although extensive clinical valida-
nant lesion post-Doppler, for a contrast-enhanced tion is needed:
power Doppler US diagnostic accuracy of 77%, as
compared with 46% for contrast-enhanced CT, and Type 1: Simple cyst. Benign, with no malignant
86% for MR imaging. potential. No further work-up.
Gray-scale contrast imaging may improve this Type 2: Few thin septations or small amount of
diagnostic accuracy even further, because significant peripheral calcification. Small chance of ma-
gray-scale parenchymal enhancement can be seen in lignancy. Evaluate cyst enhancement with
vessels too small to resolve with power Doppler. It is UCA. If no enhancement, no further follow-up

Fig. 4. A 74-year-old woman with renal failure and focal renal lesion on noncontrast CT for abdominal fullness. (A) Axial
noncontrast CT of the left kidney shows an atrophic kidney with hyperdense round, exophytic lesion (arrowheads). (B)
Longitudinal postcontrast image of the left kidney demonstrates a well-marginated anechoic structure in the mid-portion,
consistent with simple renal cyst (arrows). Note the small amount of enhancement of the renal parenchyma (calipers) consistent
with the patient’s renal failure.
M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978 969

is needed. If substantial enhancement seen, with CT, then follow with US. If substantial
assess with CT. Even if CT is negative for enhancement is seen, consider surgery.
enhancement, consider US follow-up.
Type 3: Many thin septations or several thicker Fig. 4 shows a hyperdense cyst on noncontrast CT
septations or small mural nodule. Intermediate in a patient with progressive renal failure whose
chance of malignancy. Evaluate cyst enhance- physician was unwilling to give him iodinated con-
ment with UCA. If no enhancement seen, trast. A simple cyst was seen at US, with good through
assess with CT, then follow with US. If sub- sound transmission. No enhancement was seen with
stantial enhancement is seen, consider sur- US contrast (renal cyst type 1). Fig. 5 shows a small
gery. Follow-up is mandatory if surgery is mural nodule with substantial enhancement postcon-
not performed. trast (renal cyst type 3). Pathology showed a fibrous
Type 4: Many thick septations, large mural nodule corresponding to the enhancing mural nodule,
nodule, or mural nodularity. High chance of without evidence of malignancy. This case illustrates
malignancy. Evaluate enhancement with UCA. the fact that not only neoplasms have vascularity, a
If no sonographic enhancement seen, assess source of false-positive examinations also found in

Fig. 5. A 54-year-old woman with abdominal pain. (A) Conventional longitudinal gray-scale image of the lower pole of the kidney
demonstrates a small complex cortical lesion (arrows). (B) No vascularity is identified by power Doppler in the same region
(arrow). (C) Postcontrast high mechanical index image shows an enhancing nodule (arrowheads) within the lesion. On pathology,
fibrosis with hemosiderin-laden macrophages was present within the nodule, but no malignancy was present in the cyst.
970 M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978

Fig. 6. An 80-year-old woman with abdominal pain. (A) Conventional longitudinal gray-scale image of the kidney shows a large
cystic renal lesion with numerous septations, consistent with a Bosniak III lesion (arrows). (B) Postcontrast high mechanical
index image in the same region clearly shows enhancement of the thickened septations (arrowheads) within the cystic mass,
consistent with renal cell carcinoma.

both CT and MR imaging. Another renal cyst, type 3, With US contrast, a gray-scale image is produced
is shown in Fig. 6, with multiple septations without that is similar to the cortical-medullary phase of
flow on conventional color and power Doppler, which contrast at CT, MR imaging, and angiography. The
enhanced with US contrast. Fig. 7 shows a cystic renal cortex brightly enhances and appears echogenic. The
mass with little demonstrable power Doppler flow pyramids are less echogenic than the cortex, because
precontrast, but substantial enhancement on gray scale they have less blood flow. Visualization of normal-
postcontrast. This mass is a type 4 renal cyst and is a appearing smoothly branching vessels from the renal
neoplasm until proved otherwise. hilum to the periphery is normal, and is a very useful
sign that no mass is present [6]. Findings suspicious
Pseudotumors for malignancy in a mass include vessels with an
There are several normal variants that can cause abnormal course or branching pattern on gray-scale or
difficulty in the goal of finding small renal cell power Doppler imaging. Other imaging findings that
carcinomas. A prominent column of Bertin, persistent raise the suspicion of a malignancy in a mass include
fetal lobulation, and a dromedary or splenic hump can an increase or decrease in vascularity with respect to
be mistaken for a mass, especially by the inexpe- adjacent normal cortex. A recent small series of four
rienced examiner. The morphology of the area of the patients with renal mass versus pseudotumor was
potential renal mass is important. A prominent col- evaluated with power Doppler US and Levovist.
umn of Bertin can be distinguished from a mass at Three cases of renal pseudotumors (two columns of
gray-scale US by assessing the cortical thickness with Bertin, one persistent fetal lobulation) and one case of
respect to the renal pyramid. Typically, the renal centrally located renal cell carcinoma were diagnosed
pyramid is located more deeply in the renal sinus at US, and verified with CT or surgery [32].
than usual, flanked by normal cortex. The surface of Ultrasound contrast can be used to assess renal
the kidney in a prominent column of Bertin is vascularity, cortical thickness, and pyramid spacing,
typically smooth, rather than bulging as with a renal and can be of particular usefulness to the inexpe-
cortical mass [31]. The echogenicity of a column rienced examiner when there is a suggestion of a
of Bertin pseudotumor may be slightly increased renal mass. It is likely that after this approach is
because of anisotropic effect [31]. Persistent fetal validated in a larger number of patients, renal contrast
lobulation refers to surface lobularity without an US will be the technique of choice to exclude a renal
alteration in the spacing of renal pyramids, or thick- pseudotumor in the course of normal daily practice,
ness of the renal cortex. instead of the more costly CT or MR imaging.
M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978 971

Fig. 7. A 54-year-old man with renal lesions discovered on chest CT for small pulmonary nodules. (A) Conventional longitudinal
gray-scale image of the kidney (calipers) shows large complex cystic mass (arrows). (B) No vascularity in the lesion is seen on
power Doppler ultrasound (arrows). (C) Postcontrast high mechanical index technique shows multiple areas of irregular
enhancing septations within the cystic lesion (arrowheads). Clear cell renal cell carcinoma was confirmed by pathology.

Indeterminate renal masses (28%). Another important finding was an increase in


The problem of a renal lesion that is too small to diagnostic confidence in 64% of cases. The authors
be characterized adequately on CT or MR imaging is preferred gray-scale low MI imaging techniques in
a common one. Another not infrequent clinical prob- most cases, often combined with interval-delay high
lem is the patient with a renal lesion that enhances MI images for best depiction of the abnormality, over
only minimally on CT, below the cutoff for definite power or color Doppler [34].
renal enhancement consistent with a renal cell carci- Optimal care suggests that the patient leave the US
noma. Pseudoenhancement is a potential cause of a laboratory with a definitive accurate diagnosis, rather
false-positive CT [33]. It is useful to be able to make than undergo uncertainty regarding a possible abnor-
a definitive diagnosis in such patients, rather than mality, and the additional time and expense of other
have the patient and physician endure uncertainty, modalities. Fig. 8 shows a renal mass that was inde-
long follow-ups, and additional costly testing. terminate on a dedicated CT with and without intra-
In the authors’ experience with 69 patients with venous contrast, with a measured enhancement of
indeterminate renal masses, additional information 8 HU. A prior CT scan had shown more lesion
was obtained with an US contrast-enhanced study enhancement (but still indeterminate), and this lesion
in 96% of patients. These included improved conspi- was being followed for interval growth. US with
cuity of the abnormality (51%); improved delineation contrast showed definite enhancement of both septa-
of the abnormality (41%); and exclusion of pathology tions and mural nodules, consistent with a renal cell
972 M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978

Fig. 8. A 42-year-old woman with indeterminate lesion on CT performed for abdominal pain. (A) Precontrast CT image of the
kidney has a normal appearance without focal mass. (B) Postcontrast CT of the same region shows a focal lesion (arrows) that is
hypodense relative to the adjacent renal parenchyma. The enhancement of the lesion measured 8 HU (indeterminate for
malignancy). (C ) Conventional longitudinal gray-scale ultrasound of the lower renal pole has mild heterogeneity without definite
focal lesion. (D) Focused gray-scale evaluation of the lower pole in transverse plane shows a heterogenous focal renal mass
(calipers). (E ) Postcontrast longitudinal low mechanical index image of the same region demonstrates a complex renal lesion
with enhancement of numerous septations and mural nodules (arrowheads), consistent with a renal cell carcinoma.
M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978 973

carcinoma. It is surprising to see this degree of en- should not affect the use of a noninvasive test for
hancement on an US contrast study, and yet have no detecting renovascular hypertension.
change in Hounsfield units on an optimally performed The use of UCAs to increase the number of
thin-section CT because of partial volume averaging. reflectors in the main and intrarenal arteries is an
excellent use of microbubbles. Spectral Doppler gate
placement accuracy and angle correction are im-
Renal artery stenosis proved with better vessel delineation. The ability to
improve the spectral Doppler signal by better vessel
Renal artery stenosis occurs in approximately 1% visualization is useful not only in the main renal
to 2% of hypertensive patients. Although it is a artery but also in the segmental intrarenal arteries.
relatively uncommon cause of hypertension, angio- Evaluation of the intrarenal arteries is necessary when
plasty or surgery can virtually eliminate the need for the main renal arteries cannot be visualized in their
medication to control blood pressure in some patients. entirety [44]. Although there is controversy regarding
Screening patients so that there is a higher pretest which should be studied, the main renal arteries or the
probability of the disease still leaves a high percentage intrarenal arteries, it is usually preferable to directly
of patients with negative angiograms. Angiography visualize the abnormality rather than to infer its pres-
has a small but real morbidity, coupled with the ence from an indirect test.
potentially nephrotoxic effects of iodinated contrast. Ultrasound contrast agents have been found to
Captopril nuclear renography is commonly used; increase the number of technically adequate diagnos-
however, it may miss bilateral disease [35,36]. It is tic examinations of the main renal artery [7]. UCAs
also not sensitive in those patients with limited renal have also been found to improve technical success
function, a common problem in this patient population and increase diagnostic confidence after renal artery
[37]. Although there is substantial development in angioplasty and stenting [45]. A significant difference
using CT and MR imaging angiography for renal in the enhancement curve of kidneys with renal artery
artery stenosis detection, there is no completely satis- stenosis compared with normal kidneys was found by
factory screening study [38 – 41]. Lencioni et al [46]. UCAs also can decrease the time
The sonographic detection of renal artery stenosis needed for the examination.
could be one of the commonly performed studies in a It can be argued that the renal artery stenosis ex-
sonologist’s armamentarium were it not for the deep amination should be performed only after a UCA in-
location of the main renal arteries and the presence of jection, rather than UCAs used as a rescue agent, after
accessory renal arteries. The deep horizontal course failure to see the main renal arteries. Undoubtedly,
of the renal arteries under a large amount of bowel there are some patients whose main renal arteries
limits the number of main renal arteries that can be would have been seen well without a UCA. In an ex-
seen in their entirety, especially in obese patients. amination whose technical failure rate can reach 20%
UCAs have been used recently to improve visualiza- [47] even in experienced hands, however, any tool to
tion of the main renal arteries in difficult patients. increase the accuracy and speed of the examination
Melany et al [7] found that UCAs improved main should be used routinely, rather than selectively.
renal artery visualization substantially. Two of eight
cases of stenosis were only seen postcontrast.
Interestingly, they also had seven kidneys with Renal perfusion
accessory renal arteries whose accessory arteries were
only seen postcontrast [7]. An accessory renal artery The lack of a clinically available contrast agent
occurs in up to 30% of the population [42], and severely hampers the ability of US to detect changes
theoretically can be the cause for patients’ hyperten- in renal perfusion, except in a gross fashion. Often,
sion. Increased ability to detect accessory renal arteries only a subtle alteration in renal echotexture is seen at
with UCAs may negate one of the theoretical reasons US despite a substantial renal perfusion abnormality
that the US examination has never been widely per- because of renal infarction, infection, or transplant
formed. It has been reasoned that there is no point in rejection. CT, MR imaging, or radionuclide scintig-
using US for stenosis detection because of poor raphy is usually performed to assess perfusion.
accessory renal artery detection. A recent article from
Bude et al [43], however, found that the incidence of a Infarction
hemodynamically significant stenosis isolated to an
accessory renal artery is very small (1.5%). They con- Contrast-enhanced US seems promising in the
cluded that failure to detect accessory renal arteries detection of decreased renal vascularity from infarcts.
974 M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978

It has been found to be an effective way of depicting arterial or venous thrombus. Complete sonographic
focal renal perfusion defects in the pig and rabbit imaging of the renal infarct without the added time to
[48,49]. Animal studies have been extended to the diagnosis, additional expense of other modalities,
human by Yücel et al [50], who demonstrated sub- and lack of potential nephrotoxicity found with
stantial improvement in visualization of renal infarcts iodinated contrast agents is attractive, if borne out
over power Doppler US in three patients [50]. This in larger studies.
technique may be a useful front-line study in the
radiologist’s armamentarium, especially in the patient Pyelonephritis
with compromised renal function. UCAs also may be
used to image the main renal artery and vein of these Ultrasound is commonly used as the first moda-
patients, to evaluate for potential extension of either lity for renal evaluation in cases of suspected pyelo-

Fig. 9. A 20-year-old woman with fever and flank pain. (A) Conventional longitudinal gray-scale ultrasound of the kidney
(calipers) demonstrates thickening of the renal cortex in the mid-portion without substantial gray-scale changes. (B) Longitudinal
power Doppler image in the same region shows abnormal hypoperfusion of the renal vasculature in the region of cortical
thickening (arrows). (C ) Postcontrast transverse image using low mechanical index technique demonstrates a focal hypoechoic,
or avascular, region in the cortex (arrowheads). (D) Axial CT image of the same region confirms focal hypoenhancement of the
parenchyma (arrows), consistent with pyelonephritis.
M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978 975

nephritis. After appropriate antibiotic treatment, if the use of US contrast in the detection of renal parenchy-
patient is still febrile and has significant flank pain, a mal injury in humans somewhat doubtful. The use of
CT is obtained to exclude a drainable renal abscess. UCAs may be restricted to those trauma patients with
Power Doppler has been shown to be useful in the hematuria who are otherwise asymptomatic.
detection of perfusion abnormalities associated with
pyelonephritis in children [51,52]. Fewer gray-scale, Intraoperative
color, and power Doppler changes are seen, however,
in the adult with pyelonephritis. This finding is likely There is growing interest in renal-sparing tech-
secondary to the increased mass of adults as com- niques as a result of increased detection of incidental,
pared with the average child, despite the increased clinically silent small renal tumors. Radiofrequency
penetration and power Doppler sensitivity present in ablation is being evaluated as an alternative therapy
current equipment. Contrast-enhanced CT may be in renal cell carcinoma patients who are not surgical
necessary to help distinguish between pyelonephritis, candidates, and as a treatment for small incidental
abscess, or infarction [53]. tumors. In this procedure an image-guided radiofre-
UCAs have been shown to be useful in detecting quency ablation needle is advanced into the tumor
changes consistent with infection or scarring in a and electrical energy is used to produce heat to kill
porcine model of experimental pyelonephritis [54]. the tumor cells [68]. It is important to ablate the entire
Contrast US may be used both to detect or exclude the tumor and a margin of surrounding tissue. Contrast-
perfusion defects seen with pyelonephritis, and assess enhanced US is useful for initial tumor localization,
for the presence of a drainable abscess [55]. In the and to detect residual tumor after ablation [69].
future, it is doubtful that CT will be needed for further Although CT can be used to guide this procedure,
assessment, because a repeat US can be performed US has several advantages, including real time needle
readily if the patient fails to respond to therapy. Fig. 9 placement and multiple scanning planes. The use of
demonstrates the decrease in regional perfusion seen in UCAs during the procedure can improve the detec-
a patient with pyelonephritis, with CT correlation. tion of residual tumor at the time of the procedure
(RGB, personal communication, 2003). Usually, addi-
Perfusion tional contrast cannot be given after a CT-guided
radiofrequency ablation because of nephrotoxicity
Assessment of renal blood flow can be useful in limitations and residual contrast within the kidney.
patients with both acute and chronic renal failure. This is in contradistinction to US, where small bol-
Clinically, most interest focuses on the renal trans- uses of a UCA can be given multiple times without
plant rather than the native kidney. Assessment of consideration of nephrotoxicity.
blood flow changes in the transplant can be helpful in
sorting among diagnostic possibilities for renal dys-
function, including acute tubular necrosis, acute and Summary
chronic rejection, cyclosporine toxicity, and renal
artery stenosis. Preliminary work has been performed The application of UCAs to the kidney is still in its
in a variety of experimental animals and in the human infancy; however, there are several areas of great
[56 – 63]. Solving the clinical problem of noninva- promise. UCAs may replace CT in complex renal cyst
sively determining blood flow volume also may be of evaluation and follow-up, eliminating the need for
use in other organs, such as the liver. costly CT scans with their attendant potential contrast
nephrotoxicity. This approach may decrease patient
Trauma and physician uncertainty and improve diagnostic
confidence. The use of UCAs is likely to be clinically
The sonographic detection of renal contusions, useful in the evaluation of the indeterminate small
lacerations, and hemorrhage posttrauma is less sensi- renal mass on CT or MR imaging. Another probable
tive than CT [64], in large part because of the lack of a useful application will be in renal artery stenosis.
contrast agent. With a UCA, parenchymal renal injury Routine application of UCAs may increase the per-
detection sensitivity may approach that of CT in the centage of diagnostic examinations, increase diagnos-
patient who is not morbidly obese. The use of UCAs tic confidence, and decrease examination times. It also
in the sonographic detection of acute parenchymal will likely become the first line of evaluation in
injury has been evaluated in the pig, rabbit, and dog pyelonephritis, and be useful in immediate assessment
[65 – 67]. The increasing frequency of CT scanners in of residual tumor after radiofrequency ablation. Of
emergency departments, however, makes the routine course, substantial additional work needs to be per-
976 M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978

formed in large groups of patients to prove this [11] Greenlee RT, Murray T, Bolden S, Wingo PH. Cancer
currently optimistic outlook. statistics, 2000. CA Cancer J Clin 2000;50:7 – 33.
[12] Hellsten S, Johnsen J, Berge T, Linell F. Clinically
unrecognized renal cell carcinoma. Eur Urol 1990;
18(suppl 2):2 – 3.
Acknowledgments [13] Guinan PD, Vogelzang NJ, Fremgen AM, Chmiel JS,
Sylvester JL, Sener SF, et al. Renal cell carcinoma:
The authors gratefully acknowledge research tumor size, stage and survival. J Urol 1995;153:901 – 3.
support from Bristol Myers Squibb (North Bellerica, [14] McNichols DW, Segura JW, DeWeerd JH. Renal cell
MA), and equipment and technical support from carcinoma: long-term survival and late recurrence.
Acuson, a Siemens company (Mountain View, CA), J Urol 1981;126:17 – 23.
[15] Dinney CPN, Awad SA, Gajewski JB, Belitsky P, Lan-
and ATL, Phillips Medical Systems (Bothell, WA).
non SG, Mack FG, et al. Analysis of imaging modali-
The authors thank Lisa Nelson, RN, BSN; Cynthia L. ties, staging systems, and prognostic indicators for
Peterson, BS, RDMS, RVT; Michael Clements, BS, renal cell carcinoma. Urology 1992;39:122 – 9.
RDMS, RVT; Carl Abts, RDMS; and Al Hester, [16] Tsui KH, Shvarts O, Smith RB, Figlin RA, DeKernion
RDMS, RVT for their invaluable assistance in our JB, Belldegrun A. Prognostic Indicators for renal cell
ultrasound contrast research programs. The authors carcinoma: a multivariate analysis of 643 patients
also thank Trish Dobbs for her assistance in manu- using the revised 1997 TNM staging criteria. J Urol
script preparation, and Anthony Zagar for photo- 2000;163:1090 – 5.
graphic assistance. [17] Scher HI, Motzer RJ. Bladder and renal cell carcinoma.
In: Braunwald E, Fauci AS, Kasper DL, Hauser SL,
Longo D, Jameson JL, editors. Harrison’s principles
of internal medicine. New York: McGraw-Hill; 1999.
References p. 604 – 7.
[18] Davidson AJ, Hayes WS, Hartman DS, McCarthy
[1] Robbin ML. Ultrasound contrast agents: a promising WF, Davis Jr CJ. Renal oncocytoma and carcinoma:
future. Radiol Clin North Am 2001;39:399 – 414. failure of differentiation with CT. Radiology 1993;186:
[2] Correas JM, Hélénon O, Moreau JF. Contrast-en- 693 – 6.
hanced ultrasonography of native and transplanted kid- [19] Reichelt O, Wunderlich H, Weirich T, Schlichter A,
ney diseases. Eur Radiol 1999;9(suppl 3):S394 – 400. Schubert J. Computerized contrast angiosonography: a
[3] Gramiak R, Shah PM. Echocardiography of the aortic new diagnostic tool for the urologist. BJU Int 2001;
root. Invest Radiol 1968;3:356 – 66. 88:9 – 14.
[4] Robbin ML, Eisenfeld AM. Perflenapent emulsion: [20] Atkins MB, Garnick MB. Renal neoplasia. In: Brenner
a US contrast agent for diagnostic radiology - multi- BM, editor. Brenner and Rector’s the kidney. 6th edi-
center, double-blind comparison with a placebo. Radi- tion. Philadelphia: WB Saunders; 2000. p. 1044 – 68.
ology 1998;207:717 – 22. [21] Bosniak MA. The small (V 3.0 cm) renal parenchy-
[5] Lopez-Ben R, Robbin ML, Weber TM, Smith JK, Nee- mal tumor: detection, diagnosis, and controversies.
dleman L, Berland LL. Doppler sonographic enhance- Radiology 1991;179:307 – 17.
ment of hepatic hemangiomas and hepatocellular [22] Jamis-Dow CA, Choyke PL, Jennings SB, Linehan
carcinomas after perflenapent emulsion: preliminary WM, Thakore KN, Walther MM. Small (V 3cm) renal
study. J Ultrasound Med 1999;18:109 – 16. masses: detection with CT versus US and pathologic
[6] Robbin ML. The promise of sonographic contrast correlation. Radiology 1996;198:785 – 8.
agents in the kidney. In: Nanda NC, Schlief R, Gold- [23] Forman HP, Middleton WD, Melson GL, McDlennan
berg BB, editors. Advances in echo imaging using BL. Hyperechoic renal cell carcinomas: increase in
contrast enhancement. 2nd edition. The Netherlands: detection at US. Radiology 1993;188:431 – 4.
Kluwer Academic Publishers; 1997. p. 525 – 41. [24] Yamashita Y, Ueno S, Makita O, Ogata I, Hatanaka Y,
[7] Melany ML, Grant EG, Duerinckx AJ, Watts TM, Watanabe O, et al. Hyperechoic renal tumors: anechoic
Levine BS. Ability of a phase shift US contrast agent rim and intratumoral cysts in US differentiation of
to improve imaging of the main renal arteries. Radi- renal cell carcinoma from angiomyolipoma. Radiology
ology 1997;205:147 – 52. 1993;188:179 – 82.
[8] Lencioni R, Cioni D, Bartolozzi C. Tissue harmonic [25] Siegel CL, Middleton WD, Teefey SA, McClennan
and contrast-specific imaging: back to gray scale in BL. Angiomyolipoma and renal cell carcinoma: US
ultrasound. Eur Radiol 2002;12:151 – 65. differentiation. Radiology 1996;198:789 – 93.
[9] Burns PN. Harmonic imaging with ultrasound contrast [26] Hélénon O, Merran S, Paraf F, Melki P, Correas JM,
agents. Clin Radiol 1996;51(suppl 1):50 – 5. Chrétien Y, et al. Unusual fat-containing tumors of the
[10] Goldberg BB, Liu JB, Forsberg F. Ultrasound con- kidney: a diagnostic dilemma. Radiographics 1997;17:
trast agents: a review. Ultrasound Med Biol 1994;20: 129 – 44.
319 – 33. [27] Ascenti G, Zimbaro G, Mazziotti S, Gaeta M, Settineri
M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978 977

N, Scribano E. Usefulness of power Doppler and con- the renal arteries for renovascular hypertension. Radi-
trast-enhanced sonography in the differentiation of ology 2003;226:411 – 6.
hyperechoic renal masses. Abdom Imaging 2001;26: [44] Stavros AT, Parker SH, Yakes WF, Chantelois AE,
654 – 60. Burke BJ, Meyers PR, et al. Segmental stenosis of
[28] Bosniak MA. Difficulties in classifying cystic lesions the renal artery: pattern recognition of tardus and par-
of the kidney. Urol Radiol 1991;13:91 – 3. vus abnormalities with duplex sonography. Radiology
[29] Kim AY, Kim SH, Kim YJ, Lee IH. Contrast-enhanced 1992;184:487 – 92.
power Doppler sonography for differentiation of cystic [45] House MK, Dowling RJ, King P, Bourke JL, Jardine C,
renal lesions: preliminary study. J Ultrasound Med Thomson KR, et al. Doppler ultrasound (pre- and post-
1999;18:581 – 8. contrast enhancement) for detection of recurrent ste-
[30] Bosniak MA. The use of the Bosniak classification sys- nosis in stented renal arteries: preliminary results.
tem for renal cysts and cystic tumors. J Urol 1997; Australas Radiol 2000;44:36 – 40.
157:1852 – 3. [46] Lencioni R, Pinto S, Cioni D, Bartolozzi C. Contrast-
[31] Yeh HC. Some misconceptions and pitfalls in ultra- enhanced Doppler ultrasound of renal artery stenosis:
sonography. Ultrasound Quarterly 2001;17:129 – 55. prologue to a promising future. Echocardiography 1999;
[32] Ascenti G, Zimbaro G, Mazziotti S, Gaeta M, Lamber- 16(part 2):767 – 73.
to S, Scribano E. Contrast-enhanced power Doppler [47] Hélénon O, Rody F, Correas JM, Melki P, Chauveau D,
US in the diagnosis of renal pseudotumors. Eur Radiol Chretien Y, et al. Color Doppler US of renovascular
2001;11:2496 – 9. disease in native kidneys. Radiographics 1995;15:
[33] Abdulla C, Kalra MK, Saini S, Maher MM, Ahmad 833 – 54.
E, Halpern E, et al. Pseudoenhancement of simu- [48] Taylor GA, Barnewolt CE, Claudon M, Dunning PS.
lated renal cysts in a phantom using different multi- Depiction of renal perfusion defects with contrast-
detector CT scanners. AJR Am J Roentgenol 2002; enhanced harmonic sonography in a porcine model.
179:1473 – 6. AJR Am J Roentgenol 1999;173:757 – 60.
[34] Barr RG, Robbin ML, Peterson C. Definity-enhanced [49] Girard MS, Mattrey RF, Baker KG, Peterson T, Deira-
ultrasound imaging of the kidney in patients with nieh LH, Steinbach GC. Comparison of standard and
indeterminate masses: value of contrast harmonic second harmonic B-mode sonography in the detection
imaging with bolus and infusion administration. Chi- of segmental renal infarction with sonographic contrast
cago: Radiological Society of North America; 2001. in a rabbit model. J Ultrasound Med 2000;19:185 – 92.
[35] Fommei E, Volterrani D. Renal nuclear medicine. [50] Yücel C, Özdemir H, Akpek S, Gürel K, Kapucu O,
Semin Nucl Med 1995;25:183 – 94. Araç M. Renal infarct: contrast-enhanced power Dopp-
[36] Nally Jr JV. Provocative captopril testing in the diag- ler sonographic findings. J Clin Ultrasound 2001;
nosis of renovascular hypertension. Urol Clin North 29:237 – 42.
Am 1994;21:227 – 34. [51] Dacher JN, Pfister C, Monroc M. Power Doppler sono-
[37] Bourgoignie JJ, Rubbert K, Sfakianakis GN. Angioten- graphic pattern of acute pyelonephritis in children:
sin-converting enzyme-inhibited renography for the comparison with CT. AJR Am J Roentgenol 1996;
diagnosis of ischemic kidneys. Am J Kidney Dis 166:1451 – 5.
1994; 24:665 – 73. [52] Winters WD. Power Doppler sonographic evaluation
[38] Prokop M. Protocols and future directions in imaging of acute pyelonephritis in children. J Ultrasound Med
of renal artery stenosis: CT angiography. J Comput 1996;15:91 – 6.
Assist Tomogr 1999;23(suppl 1):S101 – 10. [53] Clautice-Engle T, Jeffrey Jr RB. Renal hypoperfusion:
[39] Wittenberg G, Kenn W, Tschammler A, Sandstede J, value of power Doppler imaging. AJR Am J Roent-
Hahn D. Spiral CT angiography of renal arteries: com- genol 1997;168:1227 – 31.
parison with angiography. Eur Radiol 1999;9:546 – 51. [54] Farhat W, Traubici J, Sherman C, Williams T, Babyn P,
[40] Tello R, Thomson KR, Witte D, Becker GJ, Tress BM. McLorie G. Reliability of contrast enhanced sonogra-
Standard dose Gd-DTPA dynamic MR of renal ar- phy with harmonic imaging for detecting early renal
teries. J Magn Reson Imaging 1998;8:421 – 6. scarring in experimental pyelonephritis in a porcine
[41] Lufft V, Lufft LH, Fels LM, Egbeyong-Baiyee D, model: preliminary results. J Urol 2002;168:1114 – 7.
Tusch G, Galanski M, et al. Contrast media nephropa- [55] Kim B, Lim HK, Choi MH, Woo JY, Ryu J, Kim S,
thy: intravenous CT angiography versus intraarterial et al. Detection of parenchymal abnormalities in acute
digital subtraction angiography in renal artery stenosis: pyelonephritis by pulse inversion harmonic imaging
a prospective randomized trial. Am J Kidney Dis with or without microbubble ultrasonographic contrast
2002;40:236 – 42. agent: correlation with computed tomography. J Ultra-
[42] Cochran ST, Krasny RM, Danovitch GM, Rajfer J, sound Med 2001;20:5 – 14.
Barbaric ZM, Wilkinson A, et al. Helical CT angiog- [56] Aronson S, Wiencek JB, Feinstein SB. Assessment of
raphy for examination of living renal donors. AJR Am renal blood flow with contrast ultrasonography. Anesth
J Roentgenol 1997;168:1569 – 73. Analg 1993;76:964 – 70.
[43] Bude RO, Forauer AR, Caoili EM, Nghiem HV. Is it [57] Rubin JM, Fowlkes JB, Tuthill TA, Moskalik AP, Rhee
necessary to study accessory arteries when screening RT, Adler RS, et al. Speckle decorrelation flow mea-
978 M.L. Robbin et al / Radiol Clin N Am 41 (2003) 963–978

surement with B-mode US of contrast agent flow in a affect the assessment of the contrast replenishment.
phantom and in rabbit kidney. Radiology 1999;213: Ultrasound Med Biol 2001;27:937 – 44.
429 – 37. [64] McGahan JP, Richards JR, Jones D, Gersovich EO.
[58] Verbeek XA, Willigers JM, Prinzen FW, Peschar M, Use of ultrasonography in the patient with acute renal
Ledoux LA, Hoeks AP. High-resolution functional trauma. J Ultrasound Med 1999;18:207 – 13.
imaging with ultrasound contrast agents based on RF [65] Schmiedl UP, Carter S, Martin RW, Eubank W, Winter
processing in an in vivo kidney experiment. Ultra- T, Chang PP, et al. Sonographic detection of acute
sound Med Biol 2001;27:223 – 33. parenchymal injury in an experimental porcine model
[59] Arger PH, Sehgal CM, Pugh CR, Kirchoffer JI, Kotlar of renal hemorrhage: gray-scale imaging using a sono-
EY. Evaluation of change in blood flow by contrast- graphic contrast agent. AJR Am J Roentgenol 1999;
enhanced power Doppler imaging during norepineph- 173:1289 – 94.
rine-induced renal vasoconstriction. J Ultrasound Med [66] Hochmuth A, Fleck M, Hauff P, Reinhardt M, Kos-
1999;18:843 – 51. mehl H, Hilger I, et al. First experiences in using a new
[60] Claudon M, Barnewolt CE, Taylor GA, Dunning PS, ultrasound mode and ultrasound contrast agent in the
Gobet R, Badawy AB. Renal blood flow in pigs: diagnosis of blunt renal trauma: a feasibility study in
changes depicted with contrast-enhanced harmonic US an animal model. Invest Radiol 2000;35:205 – 11.
imaging during acute urinary obstruction. Radiology [67] Liu JB, Merton DA, Goldberg BB, Rawool NM, Shi
1999;212:725 – 31. WT, Forsberg F. Contrast-enhanced two and three di-
[61] Wei K, Le E, Bin JP, Coggins M, Thorpe J, Kaul S. mensional sonography for evaluation of intra-abdomi-
Quantification of renal blood flow with contrast- nal hemorrhage. J Ultrasound Med 2002;21:161 – 9.
enhanced ultrasound. J Am Coll Cardiol 2001;37: [68] Ogan K, Jacomides L, Dolmatch BL, Rivera FJ, Dellaria
1135 – 40. MF, Josephs SC, et al. Percutaneous radiofrequency
[62] Hosotani Y, Takahashi N, Kiyomoto H, Ohmori K, ablation of renal tumors: technique, limitations, and
Hitomi H, Fujioka H, et al. A new method for evalua- morbidity. Urology 2002;60:954 – 8.
tion of split renal cortical blood flow with contrast [69] Solbiati L, Goldberg SN, Ierace T, Dellanoce M, Livra-
echography. Hypertens Res 2002;25:77 – 83. ghi T, Gazelle GS. Radio-frequency ablation of hepatic
[63] Schlosser T, Pohl C, Veltmann C, Lohmaier S, Goene- metastases: postprocedural assessment with a US micro-
chea J, Ehlgen A, et al. Feasibility of the flash-replen- bubble contrast agent: early experience. Radiology
ishment concept in renal tissue: which parameters 1999;211:643 – 9.
Radiol Clin N Am 41 (2003) 979 – 999

CT evaluation of urinary lithiasis


Philip J. Kenney, MD
GU Radiology Section, Department of Radiology, JT N370, University of Alabama at Birmingham,
619 South 19th Street, Birmingham, AL 35233, USA

Diagnostic imaging has been a key part of the CT should have been done [5]. (Note the American
evaluation of urinary lithiasis nearly since the dis- College of Radiology still lists IVU and unenhanced
covery of the x-ray. The first radiograph of a renal CT as equivalent in their appropriateness criteria [6].)
calculus was obtained within months of Roentgen’s
report [1]. With the development of intravenous con-
trast agents by Swick in 1929, the intravenous uro- Advantages and disadvantages of CT
gram (IVU) became the unquestioned mainstay of
this evaluation (Fig. 1) [2]. In 1985 Pollack in a In the initial reports by Smith et al [4,7] and
monograph entitled Current Status of Excretory Urog- subsequent reports, the clear advantage of unen-
raphy stated ‘‘Of the various radiologic studies that hanced CT was its unsurpassed accuracy, Smith
may be used in investigating patients with flank pain initially reporting 97% sensitivity and 96% speci-
or renal colic, the excretory urogram is the most ficity in 292 patients. Fielding et al [8] reported 98%
expedient. . .its sensitivity. . .makes it invaluable’’ sensitivity and 100% specificity. Chen and Zagoria
[2]. It was often stated during the IVU period from [9] reported 100% sensitivity and 94% specificity.
1929 to 1994 that 90% of urinary calculi were Smith’s initial report in fact used nonspiral CT, but
radiopaque, proving the validity of the urogram. It quickly the use of spiral CT added the advantage of
was never proved, however, that 90% of calculi are speed. Niall et al [10] reported sensitivity of 100%
visible on conventional radiographs. Rather, Herring and specificity of 92%, with the mean time of
in 1962 reported that 90% of 10,000 calculi contained performance of CT at 4 minutes versus 63 minutes
some calcium [3]. In fact, only about 60% of urinary for IVU. The entire abdomen can be scanned literally
calculi are visible on radiographs (Fig. 2) [3]. within seconds with current multidetector CT devices
The first report of the use of unenhanced CT for (Figs. 3, 4).
detection of urinary tract calculi was by Smith et al Another advantage of CT is its ability to detect
[4] in 1994 at the annual meeting of the Radiologic nongenitourinary (GU) and nonstone disease, which
Society of North America. A spate of publications may be the cause of pain (Fig. 5). Smith et al [7] found
ensued, all showing high sensitivity of CT for detec- alternate diagnoses in 30 of 210 patients, whereas
tion of renal and ureteral calculi, with no paper since Fielding et al [8] reported nonstone GU pathology in
reporting better accuracy for any other imaging test. 14% and non-GU disease in 11% of 100 patients.
After some initial skepticism, unenhanced CT has Such abnormalities as appendicitis, diverticulitis, ad-
become the standard method for evaluating flank pain nexal disease, gallbladder disease, biliary disease, and
and suspected urinary lithiasis. In a recent case of a unsuspected tumors may be seen. The possibility of
patient suffering a contrast reaction from an IVU, a detecting non-GU disease on IVU is nearly nil.
malpractice suit was brought claiming unenhanced Although initial skeptics believed that the IVU
had an advantage of demonstrating physiologic in-
formation with the degree of delayed excretion
indicating degree of obstruction, numerous investi-
E-mail address: pkenney@uabmc.edu gators have shown that findings on CT allow pre-

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00067-8
980 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

Fig. 1. Clinical information: right flank pain. (A) Scout radiograph shows small calcification in right pelvis (arrows). (B) Urogram
film with 2.5-hour delay was required to document columning of the ureter to the stone.

Fig. 2. Clinical information: left flank pain. (A) On scout of the initial study, a urogram, no stone was identified. (B) Urogram
(improperly done with compression) failed to identify the stone (in retrospect present over sacrum [arrow]). (C) Scout view for
subsequent CT shows a calcification in left pelvis, where none was before (arrow). (D) Unenhanced CT shows mild left
hydronephrosis. (E) There is left ureterectasis (arrow). ( F) The stone is at left ureterovesicle junction (UVJ).
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 981

diction of outcome and can direct management. Most potential for significantly greater radiation exposure
importantly, it has been documented that measure- with CT, and the target population is generally young
ment of stone diameter on CT is accurate, because and has a nonfatal illness.
stone size is the single most reliable indicator of Indication creep has been reported as a common
stone passage [11]. problem. In 1997 Fielding et al [13] reported 58% of
Potential disadvantages of CT include cost, radia- her patients with CT for flank pain had urinary
tion exposure, and proper patient selection. Although lithiasis. Chen et al [14] reported the initial positive
most practitioners charge for a limited CT, consider- rate of 49% dropped to 28% when re-evaluated a year
ably less than the usual CT charge, this usually is later, whereas the rate of alternate diagnoses rose
more than the charge for an IVU, perhaps three to from 16% to 49%. One could debate, however,
five times greater [12]. One can argue, however, the whether this is truly a problem, because CT has
greater accuracy and speed of CT justifies somewhat become recognized as an accurate and efficient means
higher cost. Radiation exposure is an issue. There is of diagnosis of many abdominal processes, and is

Fig. 2 (continued).
982 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

Fig. 3. Clinical information: right flank pain. (A) Unenhanced CT demonstrates marked perinephric stranding (arrowheads), mild
pyelocaliectasis, and dilatation of proximal right ureter (arrow). (B) The stone is shown in the distal right ureter (arrow).

also now the standard method to seek appendicitis pitch of 2 or more. After a scout view, scanning from
and acute abdominal other disorders. just craniad to the kidneys to just below the bladder
base can be done in one or two breathholds in most
patients depending on the CT device. It is advanta-
Technique geous to have a radiologist assess the images before
removing the patient from the scan room. At
No preparation is necessary, and because no the author’s institution, about 12% of patients are
contrast is used there are virtually no contraindica- administered intravenous contrast, either to document
tions to performance of unenhanced CT, but the that opacity definitely lies within the ureter when that
possibility of pregnancy must be addressed in is unclear; to evaluate further some abnormality, such
women. The usual method of scanning uses 120 to as a mass or possible pyelonephritis; or to search for
140 kilovolt (peak) (kV[p]) with 5-mm collimation nonstone pathology if no stone is identified in a
and pitch of 1.5:1. Pitch of 1:1 may be preferable but patient with somewhat nonspecific symptoms. If the
is not necessary; small stones may be missed with desire is to visualize the ureter, the contrast dose can

Fig. 4. Clinical information: right abdominal pain. (A) There is mild right perinephric stranding and moderate hydronephrosis.
(B) The dilated right ureter could be followed to a stone (straight arrow); note the normal appendix (curved arrow).
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 983

Fig. 5. Clinical information: right lower quadrant pain, not colicky. (A) CT done initially without intravenous contrast shows no
hydronephrosis, mild symmetric bilateral stranding. Note oral contrast administered as symptoms not clearly that of renal colic.
(B) Enhanced images show normal excretion from both kidneys. (C) Inflamed appendix (arrow) demonstrated; appendicitis
confirmed surgically.

be limited to 50 mL and an appropriate delay (4 to tially lateral to the ipsilateral gonadal vein; lower in
5 minutes) used before scanning. the abdomen the gonadal vein crosses the ureter and
Interpretation is both most efficient and probably the ureter courses medially (Fig. 7). In the pelvis the
most accurate when done electronically and on soft- ureter usually courses through mid pelvis and then
copy. If a ureteral calculus is present but not clearly anteromedially to the trigone. With experience the
evident on the scout view, it may be useful to obtain a ureter can be followed easily to the stone, especially
conventional abdominal radiograph, particularly if the if the ureter is dilated (normally about 2 to 3 mm). In
stone is larger than 4 to 5 mm or over 300 HU [15]. some 50% to 77% [16,17] of cases a confirmatory
finding is seen with soft tissue thickening 1 to 2 mm
around the stone (soft tissue rim sign) (Fig. 8),
Interpretation resulting from edema at the site of stone impaction.
This tends to be seen more often distal than proximal
Identification of a stone within the ureter is the and with smaller rather than larger stones. The
most specific diagnostic finding (Fig. 6). Scrolling specificity of the sign has been reported at 92% [16].
sequentially through the images on a workstation, the One of the more common challenges is confi-
ureter can be followed from the renal pelvis as it dently distinguishing a true ureteral stone from a
courses caudally, anterior to the psoas muscle, ini- phlebolith, which are most common in the pelvis.
984 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

Fig. 6. Clinical information: right flank pain, only few hours duration. (A) Unenhanced CT shows no significant stranding,
questionable hydronephrosis. (B) Proximal right ureter (arrow) is not dilated. (C) The nondilated ureter (straight arrow) can be
followed readily; note gonadal vein (curved arrow). (D) Small stone is present in right ureter (arrow). This was not visible on
plain radiograph. (E) Follow-up CT documents stone no longer present.

This can be most difficult when the ureter is non- [16 – 18]. Occasionally a phlebolith may have a soft
dilated, and is difficult to follow through the pelvis tissue tail (Fig. 11) extending from one surface as
(Fig. 9). Phleboliths are typically round, whereas from a comet (21% [16]), a finding not seen with
urinary calculi are often slightly angular. Only 0% ureteral stones [19]. Phleboliths often (but not al-
to 20% of phleboliths show a soft tissue rim (Fig. 10) ways) show a central lucency, whether on scout view
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 985

Fig. 7. Clinical information: vague abdominal pain, no hematuria. (A) Normal enhanced CT shows normal proximal ureters
(arrows). (B) Note nondilated ureters (straight arrows) (not yet opacified by contrast) and gonadal veins (curved arrows).
(C) Note contrast-filled ureters in usual position in upper pelvis (arrows). (D) The normal ureters (arrows) course through mid
pelvis. (E) Unenhanced CT in a different patient shows the normal unopacified ureters (arrows) in mid pelvis. ( F) The
unopacified ureters (arrows) can be followed to the UVJ.

or demonstrated with bone windows or histogram, unusual course, say secondary to surgery, diagnosis
whereas true calculi are as dense or denser at center can be difficult. Comparison with prior CT scans can
than periphery [20]. Phleboliths in the gonadal vein be helpful because phleboliths remain stable in posi-
may be misread by those not familiar with the usual tion, whereas calculi tend to move; in the end, it may
anatomic orientation (Fig. 12). If the ureter has an be necessary to inject intravenous contrast to deter-
986 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

Fig. 8. Clinical information: right flank pain and microhematuria. (A) Unenhanced CT demonstrates mild right perinephric
stranding and moderate hydronephrosis. (B) Right ureterectasis (arrow) is shown in the pelvis. (C) A slightly irregular-shaped
stone (arrow) with soft tissue rim lies just proximal to UVJ. (D) More inferiorly, a round density with no soft tissue rim is seen,
typical phlebolith.

mine absolutely if a density lies within the ureter calyces at the poles, rather than the renal pelvis.
(see Fig. 12). Swelling of the kidney is difficult to judge, because
A number of signs secondary to the obstruction there is variation in kidney size, not always sym-
can be useful (Fig. 13). In one study, hydronephrosis metric. In general the signs are subjective and it is
was present in 69% with ureteral stones, dilation of difficult to grade severity. Perinephric stranding can
the ureter in 67%, perinephric stranding in 65%, and be seen in nonobstructed kidneys for various reasons;
periureteral edema in 65% [21]. Swelling of the if it is not asymmetric and ipsilateral to the patient’s
affected kidney may also be seen. The sensitivity of symptoms it may not be significant. One cause of the
the combination of perinephric stranding and ureteral perinephric stranding is the physiologic effect of
dilation alone for diagnosis of ureteral obstruction has obstruction on the kidney, resulting in increased
been reported as 99% [22]. Presence of these sec- lymphatic flow to relieve the pressure and redirection
ondary signs then can be taken as indicators of high from hilar to capsular channels [23,24]. Actual peri-
likelihood that there is a stone such that a very careful nephric fluid can be seen because of forniceal rupture
search is made, and that if it is not certain that a (Fig. 14). The renal edema from obstruction also
density is located in the ureter, probably in fact it causes loss of the hyperdense pyramid (mildly greater
is ureteral. attenuation of medullary pyramid versus cortex com-
There are limitations with use of the secondary monly seen on unenhanced CT especially in dehy-
signs. Hydronephrosis can be overcalled because of drated patients) (Fig. 15). This can result in the pale
an extrarenal pelvis; it is more reliable to assess the kidney, where the measured attenuation of the paren-
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 987

Fig. 9. Clinical information: vague left abdominal pain. (A) Although nondilated, the left ureter (arrow) can be followed into
the pelvis. (B) The ureter (arrow) can be followed further. (C) Followed to the UVJ (arrow), no ureteral stone was identified.
(D) More inferiorly there is a density; because this is inferior to the UVJ, it is a phlebolith and contrast administration is
not necessary.

chyma on the obstructed side is 5 to 14 HU less than [29,30]. The most consistent findings predicting stone
the normal side, an objective finding [25]. passage are stone size and location. About 90% of
A number of factors may dictate the presence and stones 1 mm do pass, but less than 50% of stones
severity of the secondary signs. It has been shown over 7 mm pass [31]. Stones 5 mm or larger that are
that the degree of obstruction as estimated from located in the proximal ureter are unlikely to pass
urograms correlates well with presence and severity spontaneously, whereas distal stones even if fairly
of secondary signs [26]. It also has been shown, large most often do pass [30]. In general stones over
however, that presence of secondary signs increases 6 mm commonly require intervention [29]. From the
in frequency with greater duration of symptoms [27]. urologist’s perspective, degree of obstruction has
These competing factors may explain the controversy never been a tenet directing management [32]; rather
as to whether likelihood of stone passage can be they are stone size, which statistically predicts pas-
predicted based on secondary signs; a few papers sage, and severity of symptoms, which controls
indicate a correlation [28] but most fail to do so urgency to intervene. Several studies have docu-
988 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

Fig. 10. Clinical information: left flank pain, microhematuria. (A) Unenhanced CT demonstrates mild left perinephric stranding
and moderate hydronephrosis; the left ureter was dilated. (B) The left ureter could be followed to a small stone in pelvis, with soft
tissue rim (straight arrow). Note two adjacent phleboliths with no rim (curved arrow).

mented the accuracy of CT estimation of stone size in which is surprisingly long (see Fig. 14). A stone at
the transverse plane (although not craniocaudal) the very distal course at the ureterovesicular junction
[11,33]. may even protrude into the bladder, but is off-center,
On routine supine images, it may be difficult to be whereas a stone in the midline has passed into the
certain if a stone has already passed into the bladder, bladder. Taking images in the prone position can
or remains within the intramural course of the ureter, prove the position (Figs. 16, 17)
Even with rather large stones, urologists often
attempt conservative management with hydration
and pain management. After a stone is detected by
unenhanced CT, the question of imaging follow-up
arises. It has been shown that a stone visible on scout
view is visible and can be followed by conventional
radiographs (see Fig. 15). Whenever a radiologist
detects a ureteral calculus on unenhanced CT, know-
ing the location, he or she should review the scout
view. If the stone is visible, that should be indicated
to the clinician. Only 17% to 47% of stones are seen
on CT scout view, however, whereas 48% to 60% are
seen on conventional film screen radiograph (Fig. 18)
[34]. Some have attempted to optimize the scout
view, but still the conventional film is more sensitive.
To some degree the visibility relates to size and
composition [35]. Nearly all calculi over 5 mm and
those over 300 HU are visible on radiographs [15].
Stones not seen on scout view and very small with
attenuation less than 200 may need to be followed
with CT.
Although nearly all urinary calculi are detected
with properly done unenhanced CT, several reports
show that stones formed as concretions of crystals of
Fig. 11. Clinical information: left flank pain. Unenhanced CT protease inhibitors (eg, indinavir) are nonopaque
demonstrates phlebolith with comet tail sign (arrow); left even on CT [36,37]. Given the poor solubility of
ureteral stone was demonstrated in proximal ureter. these drugs, and the relatively high incidence of stone
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 989

Fig. 12. Clinical information: vague right abdominal pain. (A) Unenhanced CT shows no secondary signs about right
kidney. (B) Note density in retroperitoneum (straight arrow) with soft tissue structure immediately posterior to it (curved
arrow). (C) Contrast was administered for documentation; note the now enhanced ureter (arrow) is posterior to the gonadal
vein phlebolith.

formation [37,38], a presumed diagnosis may be extend the examination or recommend further evalua-
made in patients taking this medication based on tion at a later date.
symptoms and demonstration of secondary signs
even if no stone is visualized.
A more serious potential problem with the in- Radiation issues
creasing use of unenhanced CT is the limitation in
diagnosing certain disorders, which may be found Although it is generally believed that CT causes
either as an incidental finding, or perhaps as a cause more radiation exposure than IVU, the relative risk
of presenting symptoms. Many lesions cannot be depends on the technique of each examination. Some
detected readily without use of intravenous or gastro- have reported nearly equal doses from CT and IVU
intestinal contrast, including renal tumors [39]; pyelo- [40], whereas others claim CT overall causes as much
nephritis; renal infarct (Figs. 19 – 21); and non-GU increased risk of cancer in the population as cigarette
disease, such as liver tumors. Although this is poten- smoking [41]. Morin et al [42] reported CT with 5-mm
tially more likely with poor patient selection, some- collimation, pitch of 1.5:1, and 120 kV(p) resulted in a
times renal colic can result from passage of clot 0.0036-Gy uterine dose versus 0.0015 Gy for IVU.
caused by a renal lesion, and the patient may present Denton et al [43], however, reported exposure of CT as
with typical obstructive symptoms. Sometimes there 4.7 mSv versus 1.5 for IVU, but based on a three-film
is subtle evidence on unenhanced CT images, such as IVU. The author’s department’s standard IVU has
a focal bulge of the kidney or perinephric stranding included a scout kidney, ureter, and bladder; renal
without a stone. The radiologist must be aware of the tomogram and renal cone down; three postcontrast
limitations and in the appropriate situation, after tomograms; anteroposterior and oblique kidney, ure-
discussion with the referring physician, may need to ter, and bladder; and pelvis prevoid and postvoid,
990 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

Fig. 13. Clinical information: acute left flank pain 3 hours duration. (A) Note marked left perinephric stranding, and caliectasis at
upper pole (arrow). (B) The pelvis is dilated; because of the peripheral caliectasis, this is not merely extrarenal pelvis. (C) There
is left ureterectasis with periureteral stranding (arrow). (D) Left ureteral stone (arrow) is shown, with no rim.

providing considerably more radiation. Regarding sure (Fig. 22). The radiation exposure is limited by
CT, the exact dose depends on technique; reduced performing a single test slice at the thickest portion
milliampere seconds (MAS) allow detection of most of the patient’s abdomen with reduced dose (80 to
stones with lesser radiation [44], but performing 100 mA, maintaining kV 140), and assessed by the
multiple series, such as precontrast and postcontrast, radiologist for quality. The examination is then done
or rescanning with thinner collimation significantly with the lowest dose that provides adequate quality in
increases dose. the individual patient, with every attempt to perform
In practice, widespread use of unenhanced CT for only a single pass.
diagnosis of urinary lithiasis increases radiation expo-
sure, probably more on the order of two to three times
greater for the individual patient rather than five Alternatives
to six times. It should be recognized, however, that
the population of concern is relatively young and Partly because of concerns over cost and espe-
suffering from a nonmalignant disease, and every cially radiation exposure, a number of studies have
attempt should be made to limit dose and also been published evaluating the use of sonography or
perform CT only for solid clinical indications. It sonography combined with conventional radiography
remains controversial how a pregnant patient with versus unenhanced CT [45 – 48]. These have docu-
suspected urolithiasis should be examined: limited mented the much lower sensitivity of sonographic
urogram or noncontrast CT. At the author’s institu- techniques (44%, 61%, 77%, and 24%) compared
tion, the advantage of definitive, rapid diagnosis is with sensitivity of CT of 92% to 96%. In addition CT
believed to outweigh slightly greater radiation expo- is performed and interpreted more rapidly, and allows
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 991

Fig. 14. Clinical information: severe right flank pain several hours duration. (A) Unenhanced CT shows bilateral renal calculi
with fluid anterior to right kidney (arrow). (B) Fluid is also seen more inferiorly in retroperitoneum (arrow). (C) A stone is seen
possibly within bladder, but to right of midline. (D) Contrast was administered; note the stone lies at the UVJ, within intramural
course of ureter (arrow). (E) Delayed image shows extravasation of contrast indicating the fluid collection is urine.
992 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

Fig. 15. Clinical information: left flank pain. (A) Unenhanced CT demonstrates left caliectasis (arrow). Attenuation of the left
kidney was 35 Hounsfield units, of the right 25. (B) A 7  13 mm stone is lodged in the left ureter. (C) The stone is clearly
visible on CT scout view. (D) Normal unenhanced CT on a different patient demonstrates bilateral dense pyramids (arrowheads),
a finding that can be lost if there is obstruction.

for diagnosis of other abdominal disorders, particu- radiologists, urologists, and emergency department
larly gastrointestinal tract. Some investigators have physicians such that it is now the standard of practice.
suggested the use of MR imaging, which can detect It is the duty of the radiologist to be aware of proper
dilatation and is especially sensitive to secondary technique and the details of interpretation. The radiolo-
signs [49]. Although this avoids radiation exposure, gist also has a duty to be aware of the limitations of
it does not reduce cost and is less rapidly performed unenhanced CT for detection and evaluation of various
with limited detection of non-GU pathology. nonstone disorders, particularly with poor patient
selection, and to extend the examination if appropriate.
Controversies and future developments include
cost containment with care for the selection of patients.
Summary Further attempts to reduce radiation exposure should
be made. Optimal CT technique is not needed in
Unenhanced CT has been demonstrated to be the general merely to detect urinary lithiasis. A consensus
most accurate and efficient diagnostic imaging means should be developed regarding use of CT in pregnant
to evaluate urinary lithiasis, with capability of direct- patients. Further improvements in the digital scout
ing management, and has become well accepted by view would be useful for following patients.
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 993

Fig. 16. Clinical information: acute left flank pain. (A) There is mild left perinephric stranding and borderline pelviectasis on
unenhanced CT. (B) Stone is present: at UVJ or in bladder? Note it is to the left of midline. (C) Prone view documents stone is in
ureter at UVJ.
994 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

Fig. 17. Clinical information: left flank pain. (A) There is left hydronephrosis and stranding present on unenhanced CT. (B) Stone
is present: at UVJ or in bladder? Note it is in midline. (C) Prone view shows stone drops, clearly in bladder.
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 995

Fig. 18. Clinical information: left flank pain. (A) Unenhanced CT demonstrates left hydronephrosis and renal calculus. (B) The
left ureter could be followed to a less than 2 mm stone (arrow) in distal ureter; this is very likely to pass spontaneously. (C) The
stone was not visible on CT scout view nor on plain radiograph (shown).
996 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

Fig. 19. Clinical information: acute left flank pain and hematuria. (A) Unenhanced CT reveals mild left perinephric stranding and
hydronephrosis; note bulge of right kidney (arrow). (B) Small stone was demonstrated in left ureter (arrow). (C) Repeat CT after
stone passage done with images before and after intravenous contrast demonstrates the right renal lesion (arrow) is complex with
some enhancement. Laparoscopic resection of a partially cystic renal carcinoma was performed.

Fig. 20. Clinical information: 20-year old woman with right abdominal pain, no history of stones. (A) Unenhanced CT was
unremarkable. (B) Contrast-enhanced CT demonstrates patchy areas diminished enhancement typical of pyelonephritis,
documented later by cultures.
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 997

Fig. 21. Clinical information: 66-year-old man with left flank pain, microhematuria, history of mitral valve disease. (A) Un-
enhanced CT demonstrates asymmetric left perinephric stranding, but no hydronephrosis or stone shown. (B) Contrast-enhanced
CT reveals lack of enhancement of left kidney indicative of renal infarct from arterial embolus.

Fig. 22. Clinical information: 17-year-old woman, 22 weeks pregnant with right flank pain. (A) Unenhanced CT was performed
with reduced dose. Initial test image done with 80 milliampere seconds, 140 kV was adequate, showing no secondary signs. (B)
Study performed with single series with reduced dose was negative, precluding any further examinations.
998 P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999

References Smith RC. Soft-tissue ‘‘rim’’ sign in the diagnosis of


ureteral calculi with use of unenhanced helical CT.
[1] Smith RC, Varanelli M. Diagnosis and management of Radiology 1997;202:709 – 11.
acute ureterolithiasis: CT is truth. AJR Am J Roent- [17] Kawashima A, Sandler CM, Boridy IC, Takahashi N,
genol 2000;175:3 – 6. Benson GS, Goldman SM. Unenhanced helical CT of
[2] Pollack HM, Banner MP. Current status of excretory ureterolithiasis: value of the tissue rim sign. AJR Am J
urography: a premature epitaph? Urol Clin North Am Roentgenol 1997;168:997 – 1000.
1985;12:585 – 601. [18] Bell TV, Fenlon HM, Davison BD, Ahari HK, Hussain
[3] Levine JA, Neitlich J, Verga M, Dalrymple N, Smith S. Unenhanced helical CT criteria to differentiate distal
RC. Ureteral calculi in patients with flank pain: cor- ureteral calculi from pelvic phleboliths. Radiology
relation of plain radiography with unenhanced helical 1998;207:363 – 7.
CT. Radiology 1997;204:27 – 31. [19] Boridy IC, Nikolaidis P, Kawashima A, Goldman
[4] Smith RC, Rosenfield AT, Choe KA, Essenmacher KR, CM, Sandler CM. Ureterolithiasis: value of the tail
Verga M, Glickman MG, et al. Acute flank pain: com- sign in differentiating phleboliths from ureteral calculi
parison of non-contrast-enhanced CT and intravenous at nonenhanced helical CT. Radiology 1999;211:
urography. Radiology 1995;194:789 – 94. 619 – 21.
[5] Eisenberg RL, Berlin L. Malpractice issues in radiol- [20] Traubici J, Neitlich JD, Smith RC. Distinguishing pel-
ogy. When does malpractice become manslaughter? vic phleboliths from distal ureteral stones on routine
AJR Am J Roentgenol 2002;179:331 – 5. unenhanced helical CT: Is there a radiolucency center?
[6] Fritzsche P, Amis Jr ES, Bigongiari LR, Bluth EI, AJR Am J Roentgenol 1999;172:13 – 7.
Bush WH, Choyke PL, et al. Acute onset flank pain, [21] Katz DS, Lane MJ, Sommer FG. Unenhanced helical
suspicion of stone disease. ACR Appropriateness Cri- CT of ureteral stones: incidence of associated uri-
teria 2000. Radiology 2000;215:683 – 6. nary tract findings. AJR Am J Roentgenol 1996;166:
[7] Smith RC, Verga M, McCarthy S, Rosenfield AT. Di- 1319 – 22.
agnosis of acute flank pain: value of unenhanced heli- [22] Smith RC, Verga M, Dalrymple N, McCarthy S,
cal CT. AJR Am J Roentgenol 1996;166:97 – 101. Rosenfield AT. Acute ureteral obstruction: value of
[8] Fielding JR, Steele G, Fox LS, Heller H, Loughlin KR. secondary signs on helical unenhanced CT. AJR Am
Spiral computerized tomography in the evaluation of J Roentgenol 1996;167:1109 – 13.
acute flank pain: A replacement for excretory urogra- [23] Heney NM, O’Morchoe PJ, O’Morchoe CCC. The re-
phy. J Urol 1997;157:2071 – 3. nal lymphatic system during obstructed urine flow.
[9] Chen MYM, Zagoria RJ. Can noncontrast helical com- J Urol 1971;106:455 – 62.
puted tomography replace intravenous urography for [24] Holmes MJ, O’Morchoe JO, O’Morchoe CCC. Mor-
evaluation of patients with acute urinary tract colic? phology of the intrarenal lymphatic system. capsular
J Emerg Med 1999;17:299 – 303. and hilar communications. Am J Anat 1977;149:
[10] Niall O, Russell J, MacGregor R, Duncan H, Mullins J. 333 – 52.
A comparison of noncontrast computerized tomogra- [25] Georgiades CS, Moore CJ, Smith DP. Differences of
phy with excretory urography in the assessment of renal parenchymal attenuation for acutely obstructed
acute flank pain. J Urol 1999;161:534 – 7. and unobstructed kidneys on unenhanced helical CT:
[11] Olcott EW, Sommer EG, Napel S. Accuracy of detec- a useful secondary sign? AJR Am J Roentgenol 2001;
tion and measurement of renal calculi: in vitro compari- 176:965 – 8.
son of three-dimensional spiral CT, radiography, and [26] Boridy IC, Kawashima A, Goldman SM, Sandler CM.
nephrotomography. Radiology 1997;204:19 – 25. Acute ureterolithiasis: nonenhanced helical CT find-
[12] Li J, Kennedy D, Levine M, Kumar A, Mullen J. Ab- ings of perinephric edema for prediction of degree of
sent hematuria and expensive computerized tomogra- ureteral obstruction. Radiology 1999;213:663 – 7.
phy: case characteristics of emergency urolithiasis. [27] Varanelli MJ, Coll DM, Levine JA, Rosenfield AT,
J Urol 2001;165:782 – 4. Smith RC. Relationship between duration of pain and
[13] Fielding JR, Fox LA, Heller H, Seltzer ST, Tempany secondary signs of obstruction of the urinary tract on
CM, Silverman SG, et al. Spiral CT in the evaluation unenhanced helical CT. AJR Am J Roentgenol 2001;
of flank pain: overall accuracy and feature analysis. 177:325 – 30.
J Comput Assist Tomogr 1997;21:635 – 8. [28] Takahashi N, Kawashima A, Ernst RD, Boridy IC,
[14] Chen MYM, Zagoria RJ, Saunders HS, Dyer RB. Trends Goldman SM, Benson GS, et al. Ureterolithiasis: Can
in the use of unenhanced helical CT for acute urinary clinical outcome be predicted with unenhanced helical
colic. AJR Am J Roentgenol 1999;173:1447 – 50. CT? Radiology 1998;208:97 – 102.
[15] Zagoria RJ, Khatod EG, Chen MYM. Abdominal ra- [29] Boulay I, Holtz P, Foley WD, White B, Begun FP.
diography after CT reveals urinary calculi: a method to Ureteral calculi: diagnostic efficacy of helical CT and
predict usefulness of abdominal radiography on the implications for treatment of patients. AJR Am J
basis of size and CT attenuation of calculi. AJR Am Roentgenol 1999;172:1485 – 90.
J Roentgenol 2001;176:1117 – 22. [30] Fielding JR, Silverman SG, Samuel S, Zou KH, Lough-
[16] Heneghan JP, Dalrymple NC, Verga M, Rosenfield AT, lin KR. Unenhanced helical CT of ureteral stones: a
P.J. Kenney / Radiol Clin N Am 41 (2003) 979–999 999

replacement for excretory urography in planning treat- Dixon RL, Dyer RB. CT diagnosis of acute flank pain
ment. AJR Am J Roentgenol 1998;171:1051 – 3. from urolithiasis. Semin Ultrasound CT MRI 2000;
[31] Coll DM, Varanelli MJ, Smith RC. Relationship of 21:2 – 19.
spontaneous passage of ureteral calculi to stone size [41] Lautin EM, Schoenfeld A, Choudhri A. Subservience
and location as revealed by unenhanced helical CT. of excretory urography to unenhanced CT in evaluat-
AJR Am J Roentgenol 2002;178:101 – 3. ing renal colic: A good idea? Benefits and consequen-
[32] Preminger GM, Vieweg J, Leder RA, Nelson RC. Uro- ces. Radiology 1998;209:285 – 6.
lithiasis: detection and management with unenhanced [42] Morin MJ, Sourtzis S, Foley WD, Jacobson DR, Begun
spiral CT: a urologic perspective. Radiology 1998;207: FP. Letters and replies. helical CT and renal calculi.
308 – 9. AJR Am J Roentgenol 2000;174:568 – 9.
[33] Narepalam N, Sundaram CP, Boridy IC, Yan Y, Heiken [43] Denton ERE, Mackenzie A, Greenwell T, Popert R,
JP, Clayman RV. Comparison of helical computerized Rankin SC. Unenhanced helical CT for renal colic:
tomography and plain radiography for estimating uri- is the radiation dose justifiable? Clin Radiol 1999;43:
nary stone size. J Urol 2002;167:1235 – 8. 444 – 7.
[34] Jackman SV, Potter SR, Regan F, Jarrett TW. Plain ab- [44] Spielmann AL, Heneghan JP, Lee LJ, Yoshizumi T,
dominal x-ray versus computerized tomography screen- Nelson RC. Decreasing the radiation dose for renal
ing: sensitivity for stone localization after nonenhanced stone CT: a feasibility study of single- and multidetec-
spiral computerized tomography. J Urol 2000;164: tor CT. AJR Am J Roentgenol 2002;178:1058 – 62.
308 – 10. [45] Catalano O, Nunziata A, Alei F, Siani A. Suspected
[35] Tublin ME, Murphy ME, Delong DM, Tessler FN, ureteral colic: primary helical CT versus selective heli-
Kliewer MA. Conspicuity of renal calculi at un- cal CT after unenhanced radiography and sonography.
enhanced CT: effects of calculus composition and size AJR Am J Roentgenol 2002;178:379 – 87.
and CT technique. Radiology 2002;225:91 – 6. [46] Fowler KAB, Locken JA, Duchesne JH, Williamson
[36] Blake SP, McNicholas MM, Raptopoulos V. Non- MR. US for detecting renal calculi with nonenhanced
opaque crystal deposition causing ureteric obstruction CT as a reference standard. Radiology 2002;222:
in patients with HIV undergoing indinavir therapy. 109 – 13.
AJR Am J Roentgenol 1998;171:717 – 20. [47] Sheafor DH, Hertzberg BS, Freed KS, Carroll BA, Keo-
[37] Schwartz BF, Schenkman N, Armenakas NA, Stoller gan MT, Paulson EK, et al. Nonenhanced helical CT and
ML. Imaging characteristics of indinavir calculi. J Urol US in the emergency evaluation of patients with renal
1999;161:1085 – 7. colic: prospective comparison. Radiology 2000;217:
[38] Reiter WJ, Schon-Pernerstorfer H, Dorfinger K, Hof- 792 – 7.
bauer J, Marberger M. Frequency of urolithiasis in [48] Tublin ME, Dodd GD, Verdile VP. Acute renal colic:
individuals seropositive for human immunodeficiency diagnosis with duplex Doppler US. Radiology 1994;
virus treated with indinavir higher than previously as- 193:697 – 701.
sumed. J Urol 1999;161:1082 – 4. [49] Sudah M, Vanninen R, Partagen K, Heino A, Vainio
[39] Chong WK, Wysoki M, Heller LG, Zegel HG. Renal P, Ala-Opas M. MR urography in evaluation of
carcinoma presenting with flank pain: a potential draw- acute flank plain: T2-weighted sequences and gado-
back of unenhanced CT. AJR Am J Roentgenol 2000; linium-enhanced three-dimensional FLASH compared
174:667 – 9. with urography. AJR Am J Roentgenol 2001;176:
[40] Chen MYM, Scharling ES, Zagoria RJ, Bechtold RE, 105 – 12.
Radiol Clin N Am 41 (2003) 1001 – 1017

MR imaging of renal function


Ambrose J. Huang, MD, Vivian S. Lee, PhD, MD*, Henry Rusinek, PhD
Department of Radiology—MRI, New York University Medical Center, 530 First Avenue, HCC Basement,
New York, NY 10016, USA

The kidneys maintain homeostasis by filtering and (Gd-DTPA, also known as gadopentetate dimeglu-
excreting metabolic waste products, regulating acid- mine), although there are also more novel approaches,
base balance, and moderating blood pressure and such as arterial spin labeling. The paramagnetic prop-
fluid volume. Because decreasing renal function erties of gadolinium cause a decrease in the T1 and T2
accompanies renal disease, monitoring renal function relaxation times of nearby tissues and fluids. The
permits assessment of disease progression and prog- physiologic behavior of gadolinium is governed by
nosis and is used to guide patient management and the properties of the agent to which it is chelated.
therapy. Many noninvasive tests of renal function are DTPA is a substance that, like inulin, is freely filtered
commonly used, but all have their drawbacks. Serum by the glomerulus and is neither resorbed nor secreted
creatinine levels and creatinine clearance are insensi- by the renal tubules, rendering it a convenient marker
tive measures of global function and cannot supply of glomerular filtration. When bound to gadolinium,
information about individual renal function. Renal its path through the kidneys can be traced with
scintigraphy can assess renal function but provides T1-weighted MR imaging. Such imaging examina-
limited anatomic information and exposes the patient tions are referred to as ‘‘MR renography.’’
to radiation. CT and intravenous urography (IVU) can The high spatial resolution of MR imaging allows
provide functional and anatomic information, but visualization of gadolinium contrast material within
both use nephrotoxic contrast agents and also expose distinct intrarenal regions, such as the cortex, the
the patient to radiation. The newest approach to medulla, and the collecting system (Fig. 1) [1 – 3].
studying renal function uses MR imaging, which Enhancement of the cortex primarily reflects perfusion
skirts these handicaps by simultaneously offering and glomerular filtration, whereas enhancement of the
exceptional anatomic detail and functional informa- medulla and collecting system, although dependent on
tion without exposure to ionizing radiation or neph- filtration, primarily reflects the condition of the renal
rotoxic contrast agents. MR imaging is the only tubules. The improved resolution of MR imaging,
single imaging modality with the potential to deliver when compared with scintigraphy, can potentially
a comprehensive anatomic and functional examina- provide more accurate measurements of renal perfu-
tion of the kidneys with minimal risk to the patient. sion or glomerular filtration, because these are based
The MR imaging evaluation of renal function on renal cortical measurements and are not con-
typically centers on visualizing the passage of contrast founded by tubular changes. Moreover, given that
material through the kidney. The contrast agents used different diseases affect different portions of the vas-
in MR imaging are usually gadolinium chelates, such cular-nephron system, MR renography has the poten-
as gadolinium diethylenetriamine pentaacetic acid tial, unique among all noninvasive tests, to distinguish
glomerular from tubulointerstitial pathology.
This article first reviews some technical issues
surrounding functional renal MR imaging. Next is
* Corresponding author. discussed the determination of two important renal
E-mail address: vivian.lee@med.nyu.edu (V.S. Lee). functional parameters: renal blood flow (RBF) and

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00066-6
1002 A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017

tween the two [3,5 – 8]. Although simple to implement,


this approach has several limitations. First, the as-
sumed linearity does not hold at higher Gd-DTPA
concentrations, where susceptibility effects come into
play. Second, differences across patients and MR
imaging systems make results difficult to generalize.
Third, it ignores the effects of flow on relative signal
intensity ratios measured in vessels, such as the aorta.
Fig. 1. Three-dimensional MR renography in a healthy Rusinek et al [9] have proposed an alternative
volunteer (spoiled GRE sequence, TR/TE/2.2/0.8/9, matrix approach to converting MR imaging signal intensity
134 256, coronal orientation, FOV 380 mm, slab thickness to Gd-DTPA concentration that invokes two relation-
96 mm, acquisition time 3 sec). On the left are 4 of 32 images ships. The first is the linear relationship between a
from an MR renography data set acquired 18 seconds after tissue’s relaxation rate and the concentration of gado-
2 mL of Gd-DTPA were injected, showing marked aortic and linium within it.
cortical enhancement (a – d, anterior to posterior). On the
right are images from the same patient acquired (a) 0, (b) 18,
(c) 90, and (d) 240 seconds after injection of Gd-DTPA. Each 1 1
¼ þ ½Gd  R; ð1Þ
image is 1 of 32 images from a three-dimensional data set. T 1 T1V
Note enhancement of the medulla (arrows) and the ureter
(open arrow). where T1 and T1V are the observed and precontrast
relaxation times of the tissue being studied, respec-
tively, [Gd] is the concentration of gadolinium, and R
is the relaxivity of gadolinium (4.5 L/mmols).
glomerular filtration rate (GFR). Finally, specific
The second is an empirically or theoretically
applications of functional renal MR imaging in the
derived relationship between signal intensity and T1
arenas of renovascular disease (RVD), hydronephro-
that must be adjusted across patients by a multipli-
sis, and renal transplantation are explored.
cative scaling factor.

Technical issues SI ¼ k  f ðT1Þ; ð2Þ

There are several technical issues relevant to MR where SI is observed signal intensity, k is a constant
renography. Among them, the quantification of con- multiplicative factor, and f is a monotonic relation-
trast concentration from MR imaging signal intensity ship between signal intensity and the tissue’s relaxa-
measurements, gadolinium dose optimization, and tion time, T1.
image analysis issues, such as segmentation, have Given a tissue’s precontrast signal intensity and
proved quite challenging. T1 value, one can use equations 1 and 2 to derive the
concentration of gadolinium within the tissue based
Quantification of contrast on signal intensity measurements following contrast
administration. The requirement of monotonicity for f
One difficulty with Gd-DTPA – enhanced MR im- is a restatement of the fact that above a certain Gd-
aging is the complex relationship between signal DTPA concentration, susceptibility effects govern the
intensity and Gd-DTPA concentration. MR imaging effect of Gd-DTPA on surrounding tissue and prevent
signal intensity varies with pulse sequence parameters, the determination of Gd-DTPA concentration from
the studied tissues’ precontrast and postcontrast T1 signal intensity.
values, and Gd-DTPA concentration [4]; furthermore, Additional methods for directly estimating Gd-
above a certain concentration of Gd-DTPA, suscepti- DTPA concentration from MR renography images
bility effects dominate and actually decrease the signal include inversion recovery T1 mapping [10]; methods
intensity. This is in contrast to renal scintigraphy, based on the Look and Locker T1 measurement pulse
where the number of radioactive counts increases sequence (referred to as a ‘‘Look-Locker methods’’)
linearly with the concentration of 99mTc-DTPA. [11]; and other dedicated T1 measurement methods,
Most groups have used the relative signal intensity such as T1 fast acquisition relaxation mapping [12].
ratio (SI  SI0)/ SI0, where SI0 represents baseline These methods, however, are currently unable to
signal intensity, to approximate Gd-DTPA concentra- provide whole kidney imaging with sufficient tem-
tion, relying on an assumed linear relationship be- poral and spatial resolution for MR renography.
A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017 1003

Gadolinium dose optimization sampling of the renal signal intensity curves rather
than a more desirable global assessment. Also,
Although most groups have used standard doses of whereas cortical ROI placement is relatively straight-
gadolinium for MR renography (0.1 to 0.2 mmol/kg), forward [13], satisfactory medullary ROI placement
studies suggest that lower doses may be advanta- is contingent on adequate corticomedullary differen-
geous because they avoid the susceptibility effects tiation soon after contrast injection.
that occur when gadolinium is concentrated in the To address this last problem, De Priester et al [13]
renal medulla and collecting system. Taylor et al [4] have developed an algorithm that exploits the relative
tested several combinations of two-dimensional fast ease of cortical ROI placement to quantify and
low angle shot (FLASH) pulse sequence parameters correct for the commingling of cortical and medullary
on a phantom to find an optimal sequence (repetition signal intensity, or volume averaging, thus facilitating
time [TR] = 38 milliseconds, echo time [TE] = 5 mil- the confident placement of medullary ROIs. Let the
liseconds, q = 40 degrees) for which the relationship signal intensity curve of a corticomedullary ROI,
between signal intensity and Gd-DTPA concen- CM(t), be decomposed into its cortical (C(t)) and
tration was approximately linear over a clinically medullary (M(t)) components.
relevant range of concentrations (0 to 1 mmol/L).
They tested three doses of Gd-DTPA (0.1, 0.05, and CM ðtÞ ¼ fC CðtÞ þ fM M ðtÞ
0.025 mmol/kg). When MR renography signal in-
tensity-versus-time curves (signal intensity curves, ¼ fC CðtÞ þ ð1  fC ÞM ðtÞ; ð3Þ
for short) were compared with renal scintigraphic
where fC and fM represent the cortical and medullary
time activity curves, correlation was generally poor.
fractions of the ROI, respectively.
In their study, however, the lowest dose of Gd-DTPA
Solving for M(t) yields
(0.025 mmol/kg) yielded signal intensity curves most
similar to scintigraphic time activity curves, suggest-
ing that the undesired role that susceptibility effects CM ðtÞ  fC CðtÞ
can play in MR renography is significant. This study M ðtÞ ¼ ð4Þ
1  fC
underscores the need for a clear grasp of the rela-
tionship between signal intensity and Gd-DTPA
Let time t = 0 be defined as the last time at which
concentration when conducting MR renography
the cortical ROI (and hence the corticomedullary
studies [4].
ROI) remains unenhanced. By applying similar rea-
Rusinek et al [9] used Monte Carlo simulations to
soning that was used to obtain equation 3, we get
determine optimal doses of Gd-DTPA for determin-
ing such computed functional parameters as GFR.
Like Taylor et al [4], the group found that lower CM ðtÞ  CM ð0Þ ¼
doses of Gd-DTPA were better for measurements
of renal function: 2.8 to 3.8 mL (approximately fC ðCðtÞ  Cð0ÞÞ þ ð1  fC ÞðM ðtÞ  M ð0ÞÞ ð5Þ
0.015 mmol/kg) in normal patients, and 3.8 to
5.6 mL (approximately 0.025 mmol/kg) in those with
During the period soon after contrast injection,
renal dysfunction [9]. These doses, substantially
however, the medulla has not yet enhanced, so M(t) =
lower than those typically used, have the additional
M(0). Equation 5 can be simplified, and solving for
benefit of making it possible to combine MR renog-
fC yields
raphy with a conventional contrast-enhanced MR
imaging examination in the same session.
CM ðtÞ  CM ð0Þ
Image analysis fC ¼ ð6Þ
CðtÞ  Cð0Þ

For image analysis of MR renography, functional


information is typically extracted by placing regions Equation 6 can then be substituted into equation 4
of interest (ROIs) over the cortex and medulla and to compute the medullary signal.
plotting their signal intensities over time. Image De Priester et al [13,14] tested this algorithm on
postprocessing faces several challenges. First, most five healthy volunteers and nine clinically asymp-
groups perform manual ROI analysis, which is labor- tomatic post – renal transplant patients. The authors
and time-intensive and is subject to operator errors. acquired MR renography images using a gradient
Moreover, ROI analysis usually provides only a recalled echo (GRE) sequence following administra-
1004 A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017

tion of Gd-DTPA. They compared signal intensity anatomic detail are drawbacks that limit its use.
curves generated from ROIs placed over purely Methods that have been used in MR imaging include
medullary regions in the healthy volunteers with first-pass contrast-enhanced imaging using extravas-
those calculated from the placement of corticomedul- cular gadolinium-based contrast agents, imaging us-
lary ROIs with varying cortical fractions. Although ing intravascular contrast agents, arterial spin
the algorithm was able to generate the correct general labeling techniques, and blood oxygenation level –
shape for the calculated medullary curves, there was dependent imaging.
an overestimation of their absolute signal intensities
that was more pronounced in curves generated from Renal perfusion imaging using extravascular contrast
corticomedullary ROIs with larger cortical fractions, agents
suggesting that there likely are nonenhancing struc-
tures within the cortex or medulla that are not Vallée et al [17] invoked the similar properties of
accounted for by the algorithm [13]. Gd-DTPA and 99mTc-DTPA to apply the microsphere
Boykov et al [15] have developed a semiauto- technique, which is used in nuclear medicine to
mated algorithm that aims to segment renal MR quantify RBF, to Gd-DTPA – enhanced MR imaging.
renography images into cortex, medulla, and collect- The microsphere theory relates RBF to the amount of
ing system regions based on a graph cuts approach contrast trapped in the kidney according to the
that first uses a temporal Markov model to represent following equation.
voxels as vectors of time histories of signal intensi-
ties. The algorithm then seeks a globally optimal seg- RBF contrast trapped in kidney
mentation while satisfying user-defined constraints. ¼ ð7Þ
cardiac output total contrast injected
The authors’ group tested the algorithm on simulated
MR renography data of normally and abnormally The amount of contrast trapped in the kidney is
functioning right and left kidneys subjected to vari- a theoretical amount, because unlike microspheres,
ous levels of noise and blur. Image postprocessing Gd-DTPA is not trapped in the kidney, although it is
times dramatically decreased from over 2 to 3 hours assumed that extravascular leakage of Gd-DTPA is
to roughly 8 minutes per patient. On average, over all inconsequential during the initial enhancement peri-
blur and noise levels, voxels tended to be over- od. The theoretical amount of contrast can be calcu-
classified into the cortex and the collecting system lated given the initial slopes of the renal Gd-DTPA
and underclassified into the medulla. For levels of and arterial wash-in curves and the arterial residue
blur and noise that were representative of clinical function, which is the integral of the arterial input
data, however, these segmentation errors resulted in function fitted to a gamma variate to correct for
less than 5% root-mean-square errors in the signal recirculation [18,19]. Relating the arterial integral to
intensities of these compartments [16]. Preliminary the total amount of contrast injected [20] yields an
evaluation of this algorithm looks favorable, but expression for RBF per unit volume.
testing on patients is still needed.
RBF max slopekidney
¼ ð8Þ
Renal perfusion imaging Volume max Dð1=T1Þaorta

Reduction in renal perfusion or RBF, typically This formula states that RBF per unit volume can
from renal artery stenosis (RAS), causes a decrease in be calculated using the maximum rate of contrast
GFR and can ultimately lead to permanent renal uptake by the kidney and the maximum contrast-
damage. The anatomic level and degree of RAS induced change in relaxivity in the aorta. The authors
typically used to describe it incompletely define renal converted signal intensity to T1 using an in vitro
status, because vessel diameter reductions of up to phantom-derived relationship between signal inten-
70% can occur before jeopardizing renal perfusion. sity and T1.
Evaluating renal perfusion should improve characteri- Vallée et al [17] used this method to measure
zation of RAS. Other clinical indications for the cortical and medullary blood flow in 27 patients in
assessment of renal perfusion include renal transplant three patient groups: (1) normal renal function as
dysfunction, chronic ischemic nephropathy, and drug determined by serum creatinine level, (2) RAS, and
nephropathy. In nuclear medicine, the measurement (3) renal failure. They found low cortical blood flow
of inflow of radionuclide tracers is used to determine in those patients with RAS compared with those with
renal perfusion, but exposure to radiation and poor normal renal function and low cortical and medullary
A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017 1005

blood flows in those patients with renal failure perfusion abnormalities. Furthermore, differences in
compared with those with normal renal function. these curves were more pronounced in the presence of
Values calculated were comparable with published a vasodilator such as acetylcholine, which is consistent
RBF measurements obtained using other modalities, with the results of other investigators [23,24].
including dynamic CT, 133Xe washout, and positron Other groups have reported similarly promising
emission tomography. results using different methods [25,26]. Although
these studies show that using MR imaging to quantify
Renal perfusion imaging using intravascular contrast renal perfusion is feasible, no group has validated
agents these methods against an accepted standard, and these
methods have not been commercially implemented for
Although assumptions about the limited extent widespread use. Further investigation is warranted.
of extravascular leakage of contrast during initial
enhancement may be valid, intravascular agents are Renal perfusion imaging using blood oxygenation
preferable for the measurement of renal perfusion. level – dependent imaging
Prasad et al [21] investigated renal cortical perfu-
sion imaging using an exogenous intravascular Blood oxygenation level – dependent (BOLD) im-
contrast agent, MS-325, which binds to albumin aging relies on the different magnetic properties of
after injection. The group acquired MS-325 – oxyhemoglobin and deoxyhemoglobin to achieve tis-
enhanced perfusion images with a turbo FLASH sue contrast on T2*-weighted images [27]. Although
sequence in seven pigs that underwent surgically BOLD imaging has been used to measure perfusion, its
induced RAS, and they expressed regional blood value in renal applications may reside in its capacity to
flow as the ratio of regional blood volume to indi- quantify oxygen use noninvasively. BOLD imaging is
cator mean transit time. MR imaging measurements particularly suited for assessing oxygenation of the
of renal perfusion were consistent with microsphere renal medulla, which normally functions at hypoxic
measurements, but a slight elevation of the former levels [28]. Prasad et al [27,29] used BOLD imaging to
may represent a limitation of this technique. Inter- study the effects of furosemide, acetazolamide, and
estingly, there was minimal decrease in renal per- water diuresis on renal oxygenation in 12 healthy
fusion even in the face of severe RAS, possibly volunteers. They found that furosemide decreased
related to the kidney’s innate capacity to regulate medullary R2* (1/T2*) in all patients, indicating an
blood flow. Although these results are encouraging, increase in medullary oxygenation, but it did not
intravascular contrast agents have yet to be ap- significantly affect cortical R2*. The furosemide-in-
proved for RBF measurement. duced decrease in activity, and hence oxygen use, of
proximal tubular transporters explains these findings
Renal perfusion imaging using arterial spin labeling [27]. Acetazolamide did not significantly affect corti-
techniques cal or medullary R2*. Water diuresis had similar
effects on the cortex and medulla as furosemide,
An alternative approach to measuring renal perfu- although the authors hypothesized in this case that
sion with exogenous contrast agents uses arterial spin the findings were caused by changes in regional blood
labeling techniques, and early results have been prom- flow [27].
ising. Prassad et al [22] studied the efficacy of using These preliminary studies illustrate the potential
both signal targeting with alternating radiofrequency for BOLD imaging to assess renal medullary oxy-
(STAR) angiography and STAR with echo-planar genation noninvasively; however, this technique has
imaging for readout (EPISTAR) perfusion imaging a limited capacity for determining whether changes in
to characterize surgically created RAS in pigs. oxygenation are caused by differences in blood flow
EPISTAR imaging revealed decreased signal intensi- or differences in oxygen use [30].
ties in kidneys supplied by stenotic renal arteries.
Moreover, correlating the perfusion images with con-
ventional selective angiograms showed that segmental Glomerular filtration rate
regions demonstrating reduced signal intensity were
supplied by occluded branch vessels. When the Monitoring renal function is essential in many
authors used the typical criterion of 70% stenosis by causes of acute and chronic renal insufficiency to
conventional angiography to indicate a positive study, assess prognosis, response to treatment, progression
the differences in signal intensity curves were 100% of disease, and nephrotoxicity associated with thera-
sensitive and 100% specific for the detection of renal peutic agents. Clinicians often use serum creatinine
1006 A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017

levels or creatinine clearance for this purpose, because ance of Gd-DTPA can be used to measure global GFR,
measuring them is relatively easy and safe. Serum the use of NMR spectrometers and the repeated
creatinine, however, is an insensitive indicator of renal sampling of blood and urine required preclude wide-
dysfunction; its production varies across individuals, spread clinical use.
and diet and medications can affect its kinetics [31].
Clearances of inulin and DTPA are indicators of GFR Single kidney GFR using MR relaxometry
and are better markers of renal function, but these
measurements require multiple blood and urine col- Although Gd-DTPA clearance was the basis of the
lections, and they do not provide unilateral renal global GFR studies previously mentioned, an alter-
functional information. native approach enables the measurement of single
For less invasive measurements of renal function kidney GFR based on calculations of the single
using MR imaging, early work showed that even kidney extraction fraction (EF) of a tracer like inulin
without contrast administration, the loss of cortico- or Gd-DTPA. The method is based on imaging and
medullary differentiation on T1-weighted MR imag- does not require blood or urine collection. EF is
ing indicated a serum creatinine level greater than defined as
3 mg/dL [32]. Recently, several MR imaging tech-
niques for the noninvasive measurement of single ½tracerartery  ½tracervein
kidney GFR have been developed based on the EFtracer ¼ ð10Þ
½tracerartery
imaging of the renal uptake of gadolinium chelates.
Three categories of GFR measurements using MR
imaging are reviewed: (1) global GFR determination Equation 1 relates observed T1 in a tissue to
using blood clearance of gadolinium-based contrast gadolinium concentration. Rearranging and substitut-
agents, (2) single kidney GFR determination using ing equation 1 into equation 10, we get
MR relaxometry, and (3) single kidney GFR deter-
   
mination using intrarenal kinetics. T 1precontrast T 1vein T 1artery
EFGd ¼  ð11Þ
T 1vein T 1precontrast T 1artery
Global GFR using blood clearance of
gadolinium-based contrast agents Once EF is determined, GFR can be calculated
according to the following.
In their 1992 paper, Choyke et al [33] compared
global GFR determined by clearance of 99mTcDTPA GRF ¼ EF  RBF  ð1 HctÞ; ð12Þ
(GFRTc ) with that determined by clearance of
Gd-DTPA (GFRGd) in 90 patients based on three where RBF is renal blood flow and Hct is hematocrit.
separate urine and blood samples using the stan- Three groups have published results using this
dard equation approach to measure single kidney GFR [34 – 36].
Dumoulin et al [34] tested this approach on human
½tracerurine  V volunteers and calculated EF using an inversion
GFRtracer ¼ ; ð9Þ
½tracerplasma recovery sequence to determine the T1 values of
moving blood in the renal artery and renal vein.
where V is the urine flow rate, and [tracer]x is the The EFs obtained in the study spanned a wide range
concentration of tracer in x. and were inconsistent with data published in previous
The authors calculated Gd-DTPA concentrations in clearance studies, but it was believed that clearance
urine and plasma by first determining the T1 of each studies were unsuitable for judging the EFs and GFRs
fluid using an nuclear magnetic resonance (NMR) calculated with this method.
spectrometer and then using an experimentally derived Niendorf et al [35] performed a study to validate
relationship to convert T1 into Gd-DTPA concentra- this technique against single kidney inulin clearance
tion. GFRGd correlated well with GFRTc (correlation measurements in six pigs. The inversion recovery
coefficient = 0.94), and the coefficient of variation of sequence they used to obtain T1 measurements, based
their differences was 3.6%. Ros et al [1] performed a on a Look-Locker method [11], was modified to use
similar study that combined plasma Gd-DTPA clear- GRE or EPI readout pulses for measuring relaxation
ance estimates of GFR with MR angiography and MR recovery. Several advantages resulted from this ap-
renography. Across six patients, GFRGd and GFRTc proach, including decreased sensitivity to off-reso-
correlated well (correlation coefficient = 0.98), and the nance effects, the use of large readout pulses, and
standard error was 3.85 mL/minute. Although clear- increased vessel contrast [35]. The group used the
A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017 1007

renal vein to derive renal venous T1 and the inferior constant between the two representing the rate of
vena cava to approximate renal arterial T1, and they clearance of tracer from the cortex.
measured RBF using phase-contrast flow quantifica-
tion MR imaging in the renal artery or renal vein. d½Gdm
Although GRE imaging provided better spatial reso- ¼ k  ½Gdc ; ð13Þ
dt
lution, EPI was faster and facilitated breathhold
acquisitions. EFGd and GFRGd correlated well with where [Gd]m,c are the time-varying concentrations of
EFinulin and GFRinulin (linear regression slopes for gadolinium in the medulla and cortex, respectively,
measuring GFR were 0.81 and 0.85 for GRE and EPI and k is the flow rate between compartments.
techniques, respectively), and their respective differ- In normal rats, the authors compared MR renogra-
ences were statistically insignificant. phy images acquired with a rapid acquisition with
Coulam et al [36] used a similar technique to relaxation enhancement sequence with those acquired
examine the effects of RAS on EF and GFR in a pig with a snapshot sequence, using gadolinium tetraazo-
model. MR imaging sequence modifications included cyclododecane-tetraacetate (Gd-DOTA), a glomerular
cardiac gating, adiabatic inversion, interleaved spiral contrast agent. The group converted signal intensities
readouts, and spectral-spatial excitation pulses. The to T1 values using available formulas relating signal
authors measured T1 relaxation using cardiac-gating intensity to T1 for the rapid acquisition with relaxation
during suspended respiration and approximated arte- enhancement and snapshot sequences, computed
rial input T1 using either the inferior vena cava or the Gd-DOTA concentration from T1 values using equa-
aorta, and they obtained RBF with phase-contrast tion 1, and determined k by fitting the resulting corti-
imaging of the renal arteries. EFGd and EFinulin corre- cal and medullary Gd-DOTA concentration curves to
lated well (correlation coefficient = 0.77, P < 0.01), equation 13. The snapshot sequence produced superior
although EFGd values were 22% less than EFinulin images compared with the rapid acquisition with relax-
( P = 0.01). In this study, the authors completed all ation enhancement sequence and was less affected by
calculations of EFinulin before computing EFGd, and susceptibility effects at high Gd-DOTA concentra-
they hypothesized that the additional anesthesia time tions. Using the snapshot sequence, the derived k
coupled with declining renal function adversely af- was 3.4 F 0.5 minutes1. Although plotting the initial
fected EFGd. In the kidneys with RAS, EFGd, EFinulin, rate of increase of Gd-DOTA concentration in the
RBF, and GFR were all significantly reduced. medulla against the administered dose of Gd-DOTA
This approach to measuring single kidney GFR is did indeed reveal a linear dependence, lending support
innovative, but it faces several technical challenges, to the proposed first-order kinetic model of glomerular
including difficulties estimating gadolinium concen- filtration, establishing the accuracy of this technique
tration and determining RBF using phase-contrast requires comparison to a reference standard.
flow measurements in small vessels. Laurent et al [37] used Baumann and Rudin’s [6]
first-order kinetic model and a snapshot sequence to
Single kidney GFR based on intrarenal kinetics calculate GFRGd in their study of the effects of
hypertension on renal function in rats. A few days
Gadolinium chelates traverse the kidney in a pre- after the MR imaging experiment, they compared
dictable fashion, progressing from the arterial blood to GFRinulin, calculated from the clearance of [3H]inulin
the cortex, medulla, and collecting system. This re- using equation 9, with GFRGd. GFRGd and GFRinulin
flects the normal passage of these substances through correlated well in the 17 rats studied (correlation
the nephron from the glomerular capillary through coefficient = 0.75).
Bowman’s capsule into the proximal convoluted tu- Smith et al [38] estimated GFR using a different
bule (cortex), through the loop of Henle (medulla), the two-compartment model, which was confined to the
distal convoluted tubule, the collecting duct (cortex cortex. In their model, the arteries and capillaries form
and medulla), and finally through the renal calyx. If the first compartment, and the proximal convoluted
one considers gadolinium chelates to be tracers whose tubules form the second. Two rate constants, kin and
passage through the kidney reflects the behavior of kout, describe the flow into and out of the proximal
other glomerular agents, then one can apply tracer tubules; kin represents GFR. The authors generated a
kinetic models to interpret patterns of intrarenal renal cortical Gd-DOTA concentration curve using a
enhancement in terms of parameters, such as GFR. two-dimensional cardiac-gated fast T1-weighted
Baumann and Rudin [6] proposed a first-order spoiled GRE sequence following a 0.05 mmol/kg
kinetic model of the kidney that consists of two injection of Gd-DOTA and fitted it to this model to
compartments, the cortex and medulla, and a rate estimate GFR. For the four patients studied with this
1008 A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017

Fig. 2. Multicompartmental model of the vascular-nephron system for analysis of MR renography data in terms of physiologic
parameters. Compartments of the model are intended to reflect closely the anatomy and function of the nephron. Solid arrows
indicate the passage of plasma or tracer-containing tubular fluid between compartments at flow rates Qij, whereas dotted arrows
indicate tracer-free fluid resorption, Fi, from the tubular compartments into the vasa recta. QPA = GFR. (From Lee VS, et al. Analysis
of dynamic three-dimensional (3D) MR renography: regional characterization by multicompartmental modeling. In: Proceedings of
the International Society for Magnetic Resonance in Medicine. Glasgow, Scotland, UK: 2001. p. 2059; with permission.)

approach, GFR ranged from 16 to 71 mL/min/100 g of cause [41]. As one of the few potentially curable
kidney cortex. Although these values are similar to causes of hypertension, RVD remains an important
values reported in the literature, validating this method yet challenging diagnosis. Not all patients with RAS
against established standards remains to be done. have RVD; in fact, those with essential hypertension
The authors’ group has described initial results tend to develop accelerated atherosclerosis, which
with a more expansive multicompartmental model of can lead to RAS. These diagnostic limitations have
the entire vascular-nephron system [39]. The model generated controversies surrounding treatment. Van
considers the distinct cortical and medullary functional Jaarsveld et al [42] concluded from their multicenter
nephron units (artery-capillary, proximal convoluted trial that treating hypertension secondary to RVD
tubule, loop of Henle, distal convoluted tubule, col- with balloon angioplasty was not much better than
lecting duct, and calyces-ureter) as separate compart- treating it with medicine alone, although the criterion
ments (Fig. 2). The cortex, medulla, and collecting they used for RVD (RAS causing only greater than
system are then expressed as a linear combination of 50% narrowing) may undermine their conclusions.
these compartments. A series of first-order differential Most anatomic tests, such as conventional angi-
equations models the course of Gd-DTPA through ography, MR angiography, and CT angiography, are
pairs of compartments over time. Like Smith et al’s limited in their ability to diagnose RVD because they
kin [38], the rate constant describing the passage of rely on RAS as the sole criterion. Angiotensin con-
Gd-DTPA from the arteries and capillaries to the verting enzyme – inhibitor (ACE-I) renal scintigraphy
proximal tubules represents GFR. Using a fast three- is the best predictor of response to therapy because it
dimensional MR renography technique, the authors is a functional test of renal ischemia. It does not,
implemented this model and computed GFR in a series however, supply anatomic information needed for
of nine subjects (18 kidneys) and found good correla- therapeutic planning.
tion (correlation coefficient = 0.76) with same-day When performed with MR angiography, MR
gamma camera – and blood clearance – derived mea- renography has the potential to provide an anatomic
sures of single kidney GFR using 99mTcDTPA [40]. and functional evaluation of RVD. Preliminary data
One advantage of this model over previous ones is its are encouraging, although small sample sizes and
inclusion of renal structures distal to the proximal susceptibility effects from concentrated contrast in
tubules. In principle, this model can assess tubular the medulla and collecting system as a result of
physiology and pathology based on MR renogra- standard doses of gadolinium have limited early
phy, both of which have been almost impossible to studies [1,43,44]. In a study of 10 patients, Ros et al
evaluate noninvasively. [1] used a turbo FLASH sequence with 0.05 mmol/kg
of Gd-DTPA and time-of-flight MR angiography to
diagnose RAS. They described one patient in whom
Angiotensin converting enzyme inhibitor MR severe left RAS shown by conventional angiography
renography for RVD corresponded to decreased left medullary enhance-
ment depicted on MR renography. Using 0.1 mmol/kg
Of the 60 million people with hypertension, an or more of gadolinium, Grenier et al [43] observed a
estimated 1% to 5% have RVD as the underlying band of low signal intensity on T1-weighted images
A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017 1009

that progressed centripetally over the course of more absence of captopril. Following captopril administra-
than 4 minutes. The susceptibility effects of con- tion, however, the kidney supplied by the stenotic
centrated gadolinium in the medulla and collecting renal artery demonstrated little washout of Gd-DTPA
system caused this signal loss and precluded con- during the imaging period. These results are consis-
trast quantification. tent with those of ACE-I renal scintigraphy.
The successful use of an ACE-I (such as capto- The authors’ group implemented an ACE-I MR
pril) in renal scintigraphy is the basis for its in- renography protocol combined with MR angiography
corporation into MR renography protocols [3]. in 32 patients with suspected RVD [3]. Imaging
Decreased renal perfusion pressure in patients with consisted of a two-dimensional turbo FLASH se-
RAS activates the renin-angiotensin system and quence using a 2-mL (0.013 mmol/kg) dose of
increases production of angiotensin II. Angiotensin Gd-DTPA for MR renography followed by con-
II causes vasoconstriction of the efferent glomerular trast-enhanced MR angiography using a standard
arteriole and restores renal perfusion pressure and (0.14 mmol/kg) dose of Gd-DTPA. It was found
glomerular filtration to normal or near-normal levels. that patients with elevated serum creatinine levels
This compensated RAS may not manifest any per- (
2 mg/dL) had depressed cortical and medullary
fusion or filtration abnormalities on renal scintigra- signal intensities at 1 to 4 minutes following Gd-DTPA
phy or MR renography. Administering an ACE-I injection when compared with patients with normal
lowers GFR in the setting of RVD because it blocks serum creatinine levels ( < 2 mg/dL) (Fig. 3). Effects
the production of angiotensin II, which decreases of the ACE-I on cortical and medullary enhancement
efferent glomerular arteriolar vasoconstriction and depended on the presence of RAS and the serum
reduces perfusion pressure. creatinine level. In patients with RAS, medullary
Prasad et al [45] used captopril MR renography enhancement following ACE-I administration was
to evaluate the hemodynamic significance of uni- slightly less than in those patients without RAS
lateral RAS in their porcine model. They performed ( P values ranged from 0.1 to 0.2), whereas there was
T1-weighted MR renography using a three-dimen- no such difference without ACE-I administration.
sional fast imaging with steady state precession Patients with elevated serum creatinine levels had
sequence and a 0.1 mmol/kg bolus of Gd-DTPA. depressed enhancement regardless of the presence or
The authors showed that the signal intensity curves absence of RAS and regardless of ACE-I administra-
of the right versus left kidneys differed little in the tion. This inability to further characterize RVD in

Fig. 3. Renal cortical (A) and medullary (B) relative signal intensity curves with standard error bars. Patients with serum
creatinine less than 2 mg/dL (solid line) are compared with those with elevated serum creatinine (dotted line). Decreased
medullary enhancement over 1 to 3 minutes in patients with renal insufficiency (dotted line, B) reflects less Gd-DTPA filtered at
the glomerulus and less passing into the loop of Henle (medulla). (From Lee VS, et al. MR renography with low-dose
gadopentetate dimeglumine: feasibility. Radiology 2001;221:371 – 9; with permission.)
1010 A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017

Fig. 4. Middle-aged hypertensive woman with mild left RAS by MR angiography (arrowhead). MR renography shows normal
medullary enhancement at baseline and decreased medullary enhancement following angiotensin converting enzyme inhibitor
injection, implying significant stenosis. (Modified from Lee VS, et al. MR renography with low-dose gadopentetate dime-
glumine: feasibility. Radiology 2001;221:371 – 9; with permission.)

patients with renal insufficiency represents a limita- lem. Both ultrasound and IVU primarily evaluate the
tion of this approach. Among patients with normal morphology of the urinary tract and have difficulty
serum creatinine levels, however, ACE-I administra- distinguishing dilated and obstructed urinary tracts
tion did unmask decreased GFR by depressing med- from those that are merely dilated. Diuretic renal
ullary enhancement in patients with RAS (Fig. 4). scintigraphy, despite its poor spatial resolution, can
ACE-I MR renography may even be useful in contribute functional information to the assessment,
transplant RAS. Sharma et al [46] studied the role of but the test suffers an unacceptably high false-posi-
ACE-I MR renography in 11 post – renal transplant tive rate [47]. Contrast-enhanced CT can provide
patients with hypertension. They performed ACE-I functional information and excellent anatomic infor-
MR renography using a turbo FLASH sequence and a mation, but the cost of the necessarily high radiation
2-mL dose of Gd-DTPA in conjunction with three- doses is problematic, particularly in the pediatric
dimensional phase-contrast MR angiography of the population, and the risk of nephrotoxicity associated
transplant renal arteries. In patients with less than with iodinated contrast agents is unsuitable for those
40% RAS, cortical signal intensity curves before and with renal insufficiency. Faster imaging and non-
after injection of 50 mg of captopril did not differ nephrotoxic contrast agents have rendered MR imag-
significantly. The cortical signal intensity curves of ing increasingly applicable for the evaluation of
patients with at least 40% RAS had a lower peak than genitourinary diseases in general and hydronephrosis
those of patients with less than 40% RAS; captopril in particular [8,48 – 50]. In addition, MR imaging
exaggerated these differences. eliminates the exposure to ionizing radiation and
These results illustrate the promise of ACE-I MR nephrotoxic agents inherent in the other modalities.
renography for evaluating hemodynamically signifi- These imaging strategies are collectively termed
cant RAS, although whether or not this technique is ‘‘MR urography.’’ Not surprisingly, lessons learned
superior to conventional anatomic studies in its from interpreting IVU or CT studies readily apply to
ability to predict a response to revascularization the interpretation of MR imaging studies (Fig. 5).
remains debatable. Wen et al [7] studied the effectiveness of Gd-
DTPA – enhanced MR imaging in assessing the func-
tion of three classes of rat kidneys: (1) nonobstructed
MR urography for hydronephrosis control, (2) partially obstructed, and (3) completely
obstructed. They acquired GRE images following the
Hydronephrosis is dilatation of the renal collect- injection of 1 mL/kg of Gd-DTPA. The partially
ing system. It may be congenital or acquired, ob- obstructed kidneys demonstrated delayed contrast
structive or nonobstructive, and clinically significant enhancement and washout when compared with the
or insignificant. The goals of the imaging evaluation normal controls. These changes were more pro-
of hydronephrosis are threefold: (1) delineation nounced in the completely obstructed kidneys, where
of the extent of dilatation of the collecting system; there was continued accumulation of contrast in the
(2) diagnosis of an obstructing cause, if any; and cortex and medulla and delayed appearance of con-
(3) evaluation of its effects on renal function. Many trast in the collecting system. These findings are
imaging modalities have been applied to this prob- analogous to the classic delayed nephrogram and
A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017 1011

Fig. 5. MR images of a 64-year-old man with a history of bladder cancer cystectomy and neobladder construction. (A) A
coronal single-shot T2-weighted image shows left-sided hydronephrosis and is suspicious for obstruction. (B) A slightly more
anterior image from the same coronal T2-weighted acquisition again shows dilatation of the renal calyces and ureterectasis.
(C) A coronal maximum intensity projection of a Gd-DTPA – enhanced three-dimensional spoiled GRE acquisition obtained
5 minutes after intravenous injection of gadolinium shows prompt excretion into the dilated collecting system, indicating
absence of functional obstruction.

delayed pyelogram observed during the IVU and CT sional inversion recovery turbo spin echo sequence,
evaluations of obstructive hydronephrosis. dynamic MR imaging using a two-dimensional
MR urography is superior to other modalities T1-weighted GRE sequence using 0.1 mmol/kg of
because it provides better anatomic and functional Gd-DTPA and 0.3 mg/kg of furosemide for distention
imaging in a single setting. Rohrschneider et al [48] of the urinary tract, and diuretic renal scintigraphy
compared MR urography with a combination of using 99mTc mercaptoacetyltriglycine and 0.5 mg/kg
ultrasound, diuretic renal scintigraphy, and IVU for of furosemide. MR urography showed the level of
the evaluation of 20 piglets that underwent surgically stenosis and the more proximal urinary tract in all
induced urinary tract obstruction. They performed of the cases, whereas IVU showed the same in only
static MR imaging using a T2-weighted three-dimen- half of the cases. Ultrasound almost never showed
1012 A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017

the level of stenosis. MR urography was also supe- for these entities on conventional MR imaging have
rior to IVU and ultrasound in demonstrating the been described [52 – 55]. Medical complications are
urinary tract distal to the stenosis. A distinct advan- diagnostically more problematic. They include acute
tage of MR urography was its ability to detect other allograft rejection; chronic allograft rejection; acute
findings, such as various fluid collections (using the tubular necrosis; cyclosporine A toxicity; infection;
static sequence) and their causes (using the dynamic and transplant-associated malignancies (lymphoma
sequence to depict contrast extravasation). The and posttransplant lymphoproliferative disorder)
authors used signal intensity curves to characterize [52]. In the early posttransplant period, acute allograft
relative renal function and urinary excretion, the rejection and acute tubular necrosis are the most
results of which agreed well with results obtained important causes of renal allograft dysfunction [2],
using diuretic renal scintigraphy. and the long-term function of the renal graft and
Katzberg et al [8] also investigated the quantita- survival of the patient depend crucially on distin-
tive analysis of MR renography data in their ana- guishing these two entities from one another, a task
tomic and functional evaluation of the kidneys of made more difficult by the possibility of their coex-
11 patients with suspected unilateral hydronephrosis. istence in the same patient.
MR renography images consisted of three 8-mm thick Imaging modalities currently used to assess renal
coronal sections acquired with a fast spoiled GRE allograft dysfunction include Doppler ultrasound and
sequence following injection of 0.05 to 0.5 mmol/kg renal scintigraphy, neither of which can differentiate
of gadolinium-based contrast. Although the small reliably among the medical causes of impaired renal
size and modest number of cases of obstructive function [56]. Definitive diagnosis of these condi-
hydronephrosis limited this study, the medullary tions often requires a renal biopsy, an invasive
enhancement pattern was delayed in hydronephrotic procedure with risks that include hemorrhage, arte-
compared with normal kidneys. Whether a qualitative riovenous fistula, pseudoaneurysm, and infection,
interpretation of MR renography requires supplemen- each of which alone can lead to loss of the renal
tation with quantitative analysis to diagnose func- allograft. The challenge is to find a reliable, non-
tional obstruction, however, remains to be established. invasive, and comprehensive method of examining
A related application is the differentiation of the renal allograft and obviate renal biopsy. Early
hydronephrosis from pyonephrosis, which is not results with functional MR imaging are promising
always straightforward. The typical diagnostic criteria and are based on visualizing distinct patterns of
used are sonographic (an anechoic calyceal system in gadolinium enhancement associated with pathologies
the former and a heterogeneously echoic calyceal that affect different portions of the nephron (Fig. 6).
system in the latter), but these are not reliable [51]. Szolar et al [2] used MR renography to examine
Conventional MR imaging also has difficulty sepa- renal allografts during the posttransplant periods of
rating the two conditions, because both often appear 23 consecutive patients with clinically suspected
hypointense on T1-weighted images and hyperintense acute allograft rejection or acute tubular necrosis
on T2-weighted images. Chan et al [51], using and demonstrated distinct patterns of cortical and
diffusion-weighted imaging to study 12 consecutive medullary enhancement in the two groups. Using
patients with pelvicaliectasis detected by ultrasound, Gd-DTPA – enhanced GRE images, 12 out of 13 pa-
found that hydronephrotic collecting systems had tients with pure acute tubular necrosis were found to
significantly higher apparent diffusion coefficients have a slight decrease and delay in cortical enhance-
than pyonephrotic collecting systems at a b-factor ment and a uniphasic instead of biphasic medullary
of 1000 s/mm2 (mean apparent diffusion coefficients enhancement pattern when compared with normal
= 2.98 F 0.65 103 mm2/s versus 0.64 F 0.35 controls. All 10 patients with acute allograft rejection
103 mm2/second, P < 0.001). had significantly decreased cortical and medullary
enhancement when compared with normal controls
and patients with acute tubular necrosis. Four patients
MR imaging for renal transplant evaluation with acute allograft rejection had superimposed acute
tubular necrosis, and MR renography was unable to
Complications following renal transplantation are distinguish them from those who had acute allograft
generally categorized as surgical or medical. Surgical rejection alone.
complications usually manifest themselves in the Although less common with newer immunosup-
immediate or early postoperative period and include pressive agents, cyclosporine A toxicity can also cause
RAS and subsequent infarction, renal vein thrombo- transplant dysfunction. Agildere et al [56] used a turbo
sis, urinary leak, and lymphocele. Typical findings FLASH sequence with 2 mL of Gd-DTPA to obtain
A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017 1013

cortical and medullary signal intensity curves for to the conspicuous role that decreased cortical perfu-
studying the differences between acute allograft rejec- sion plays in acute allograft rejection.
tion and cyclosporine A toxicity in 17 renal transplant Functional studies also have proved useful in
patients. Although the sample size was small, results diagnosing ureteral complications of renal transplan-
showed abnormally low initial mean signal intensities tation. Dörsam et al [57] evaluated the ability of
and low final mean steady-state signal intensities in MR urography to diagnose such complications in
patients with acute allograft rejection compared with 15 patients, 11 of whom had elevated serum creatinine
those with cyclosporine A toxicity, likely attributable levels. In six patients whose MR urography exami-

Fig. 6. Renal transplant dysfunction in a 62-year-old man who underwent transplantation 10 weeks earlier. Conventional
contrast-enhanced three-dimensional T1- and T2-weighted imaging of the transplanted kidney showed (A) a patent arterial
anastomosis, (B) a patent venous anastomosis, and (C) a normal collecting system without ureteral obstruction or lymphocele.
Low-dose functional MR renography (dashed line, similar to the technique shown in Fig. 1) using 4-mL Gd-DTPA revealed
(D) normal cortical perfusion, (E) slightly delayed medullary enhancement, and (F) markedly diminished contrast excretion
when compared with MR renography performed 6 days after transplantation when renal function was normal (solid line). This
pattern of enhancement suggests the diagnosis of acute tubular necrosis rather than rejection [2]. This was confirmed by biopsy.
1014 A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017

Fig. 6 (continued).

nations showed moderate hydronephrosis of the allo- renal status with minimal risk to the patient, simulta-
graft but no obstruction, subsequent clinical follow-up neously improving diagnosis while lowering medical
revealed stable performance of the allograft. costs by virtue of its being a single test [49]. The
strengths of MR imaging lie in its high spatial and
temporal resolution and its lack of exposure to
Summary ionizing radiation and nephrotoxic contrast agents.
This article reviews the use of MR imaging for
MR imaging is the only single noninvasive test quantification of renal functional parameters and its
that can potentially provide a complete picture of application to clinical problems, such as RVD, hydro-
A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017 1015

nephrosis, and renal transplantation. Although advan- [9] Rusinek H, Lee VS, Johnson G. Optimal dose of
ces in both the technical and clinical aspects of Gd-DTPA in dynamic MR studies. Magn Reson Med
functional renal MR imaging have been made, much 2001;46:312 – 6.
[10] Scheffler K, Hennig J. T1 quantification with inver-
remains to be done. The preliminary results reported
sion recovery trueFISP. Magn Reson Med 2001;45:
in the many studies reviewed are exciting, but these
720 – 3.
techniques need to be validated against accepted [11] Look DC, Locker DR. Time saving in measurement of
standards where such standards exist. In addition, NMR and EPR relaxation times. Rev Sci Instrum 1970;
and perhaps more important, the effects of these new 41:250 – 1.
diagnostic methods on patient outcomes must be [12] Chen Z, Prato FS, McKenzie C. T1 fast acquisition
studied. Finally, further progress in image processing relaxation mapping (T1-FARM): an optimized recon-
and analysis must be made to make functional renal struction. IEEE Trans Med Imaging 1998;17:155 – 60.
MR imaging truly practical. With these advances, one [13] de Priester JA, den Boer JA, Giele EL, Christiaans
can expect functional renal MR imaging to play an MH, Kessels A, Hasman A, et al. MR renography:
an algorithm for calculation and correction of cortical
ever-expanding and influential role in the care and
volume averaging in medullary renographs. J Magn
management of the patient with renal disease.
Reson Imaging 2000;12:453 – 9.
[14] de Priester JA, Kessels AG, Giele EL, den Boer JA,
Christiaans MH, Hasman A, et al. MR renography by
References semiautomated image analysis: performance in renal
transplant recipients. J Magn Reson Imaging 2001;
[1] Ros PR, Gauger J, Stoupis C, Burton SS, Mao J, 14:134 – 40.
Wilcox C, et al. Diagnosis of renal artery stenosis: [15] Boykov Y, Lee VS, Rusinek H, Bansal R. Segmenta-
feasibility of combining MR angiography, MR renog- tion of dynamic N-D data sets via graph cuts using
raphy, and gadopentetate-based measurements of glo- markov models. In: Niessen W, Viergeres M, editors.
merular filtration rate. AJR Am J Roentgenol 1995;165: Proceedings of the 4th International Conference on
1447 – 51. Medical Image Computing and Computer-Assisted In-
[2] Szolar DH, Preidler K, Ebner F, Kammerhuber F, Horn tervention. Utrecht: Springer; 2001. p. 1058 – 66.
S, Ratschek M, et al. Functional magnetic resonance [16] Huang AJ, Boykov Y, Rusinek H, Lee VS. Validation
imaging of human renal allografts during the post- of a graph cuts algorithm for semi-automated segmen-
transplant period: preliminary observations. Magn Re- tation of magnetic resonance renographic images.
son Imaging 1997;15:727 – 35. In: Proceedings of the International Society for Mag-
[3] Lee VS, Rusinek H, Johnson G, Rofsky NM, Krinsky netic Resonance in Medicine, 11th Scientific Meeting
GA, Weinreb JC. MR renography with low-dose gado- and Exhibition. Toronto: ISMRM; 2003. p. 1459.
pentetate dimeglumine: feasibility. Radiology 2001; [17] Vallée JP, Lazevras F, Khan HG, Terrier F. Absolute
221:371 – 9. renal blood flow quantification by dynamic MRI and
[4] Taylor J, Summers PE, Keevil SF, Saks AM, Diskin J, Gd-DTPA. Eur Radiol 2000;10:1245 – 52.
Hilton PJ, et al. Magnetic resonance renography: opti- [18] Thompson HK, Starmer F, Whalen RE, McIntosh HD.
misation of pulse sequence parameters and Gd-DTPA Indicator transit time considered as a gamma variate.
dose, and comparison with radionuclide renography. Circ Res 1964;14:502 – 15.
Magn Reson Imaging 1997;15:637 – 49. [19] Peters AM, Gunasekera RD, Henderson BL, Brown J,
[5] Wolf GL, Hoop B, Cannillo JA, Rogowska JA, Hal- Lavender JP, De Souza M, et al. Noninvasive measure-
pern EF. Measurement of renal transit of gadopentetate ment of blood flow and extraction fraction. Nucl Med
dimeglumine with echo-planar MR imaging. J Magn Commun 1987;8:823 – 37.
Reson Imaging 1994;4:365 – 72. [20] Lassen NA, Perl W. Tracer kinetic methods in medical
[6] Baumann D, Rudin M. Quantitative assessment of rat physiology. New York: Raven Press; 1979.
kidney function by measuring the clearance of the con- [21] Prasad PV, Cannillo J, Chavez DR, Pinchasin ES, Do-
trast agent Gd (DOTA) using dynamic MRI. Magn lan RP, Walovitch R, et al. First-pass renal perfusion
Reson Imaging 2000;18:587 – 95. imaging using MS-325, an albumin-targeted MRI con-
[7] Wen JG, Chen Y, Ringgaard S, Frokiaer J, Jorgensen trast agent. Invest Radiol 1999;34:566 – 71.
TM, Stodkilde-Jorgensen H, et al. Evaluation of renal [22] Prasad PV, Kim D, Kaiser AM, Chavez D, Gladstone
function in normal and hydronephrotic kidneys in rats S, Li W, et al. Noninvasive comprehensive characteri-
using gadolinium diethylenetetramine pentaacetic acid zation of renal artery stenosis by combination of STAR
enhanced dynamic magnetic resonance imaging. J Urol angiography and EPISTAR perfusion imaging. Magn
2000;163:1264 – 70. Reson Med 1997;38:776 – 87.
[8] Katzberg RW, Buonocore MH, Ivanovic M, Pellot-Ba- [23] Levin DC, Beckmann CF, Serur JR. Vascular resist-
rakat C, Ryan JM, Whang K, et al. Functional, dynamic, ance changes distal to progressive arterial stenosis:
and anatomic MR urography: feasibility and prelimi- a critical re-evaluation of the concept of vasodilator
nary findings. Acad Radiol 2001;8:1083 – 99. reserve. Invest Radiol 1980;15:120 – 8.
1016 A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017

[24] Shipley RE, Gregg DE. The effect of external constric- [38] Smith AM, Materne R, Van Beers BE. Quantitative
tion of blood vessel on blood flow. Am J Physiol 1944; measurement of blood perfusion, GFR and arterial
141:289 – 96. vascular fraction in the kidney cortex. In: Proceedings
[25] Berr SS, Hagspiel KD, Mai VM, Keilholz-George S, of the International Society for Magnetic Resonance in
Knight-Scott J, Christopher JM, et al. Perfusion of the Medicine. 2001.
kidney using extraslice spin tagging (EST) magnetic [39] Lee VS, Rusinek H, Kim S, Leonard E, Lee P, Johnson
resonance imaging. J Magn Reson Imaging 1999;10: G. Analysis of dynamic three-dimensional (3D) MR
886 – 91. renography: regional characterization by multicompart-
[26] Kost Jr CK, Li P, Williams DS, Jackson EK. Renal mental modeling. In: Proceedings of the International
vascular responses to angiotensin II in conscious spon- Society for Magnetic Resonance in Medicine, 9th Sci-
taneously hypertensive and normotensive rats. J Cardio- entific Meeting and Exhibition. Glasgow, Scotland,
vasc Pharmacol 1998;31:854 – 61. UK: ISMRM; 2001.
[27] Prasad PV, Edelman RR, Epstein FH. Noninvasive [40] Lee VS, Rusinek H, Huang AJ, Leonard E. Single
evaluation of intrarenal oxygenation with BOLD kidney GFR measured using 3D MR renography
MRI. Circulation 1996;94:3271 – 5. and a multicompartmental model. In: Proceedings of
[28] Brezis M, Rosen S. Hypoxia of the renal medulla – the International Society for Magnetic Resonance in
its implications for disease. N Engl J Med 1995;332: Medicine, 11th Scientific Meeting and Exhibition.
647 – 55. Toronto: ISMRM; 2003. p. 46.
[29] Prasad PV, Chen Q, Goldfarb JW, Epstein FH, Edel- [41] Pickering TG, Blumenfeld JD, Laragh JH. Renovascu-
man RR. Breath-hold R2* mapping with a multiple lar hypertension and ischemic nephropathy. In: Bren-
gradient-recalled echo sequence: application to the ner BM, editor. Brenner and Rector’s the kidney.
evaluation of intrarenal oxygenation. J Magn Reson Philadelphia: WB Saunders; 1996. p. 2106 – 25.
Imaging 1997;7:1163 – 5. [42] van Jaarsveld BC, Krijnen P, Pieterman H, Derkx FH,
[30] Prasad PV, Epstein FH. Changes in renal medullary Deinum J, Postma CT, et al. The effect of balloon
pO2 during water diuresis as evaluated by blood oxy- angioplasty on hypertension in atherosclerotic renal-
genation level-dependent magnetic resonance imaging: artery stenosis. Dutch Renal Artery Stenosis Interven-
effects of aging and cyclooxygenase inhibition. Kidney tion Cooperative Study Group. N Engl J Med 2000;
Int 1999;55:294 – 8. 342:1007 – 14.
[31] Kasiske BL, Keane WF. Laboratory assessment of [43] Grenier N, Trillaud H, Combe C, Degreze P, Jeandot
renal disease: clearance, urinalysis, and renal biopsy. R, Gosse P, et al. Diagnosis of renovascular hyper-
In: Brenner BM, editor. Brenner and Rector’s tension: feasibility of captopril- sensitized dynamic MR
the kidney. Philadelphia: WB Saunders; 1996. imaging and comparison with captopril scintigraphy.
p. 1137 – 74. AJR Am J Roentgenol 1996;166:835 – 43.
[32] Semelka RC, Corrigan K, Ascher SM, Brown JJ, Co- [44] Laissy JP, Faraggi M, Lebtahi R, Soyer P, Brillet G,
lindres RE. Renal corticomedullary differentiation: ob- Mery JP, et al. Functional evaluation of normal and
servation in patients with differing serum creatinine ischemic kidney by means of gadolinium-DOTA en-
levels. Radiology 1994;190:149 – 52. hanced TurboFLASH MR imaging: a preliminary com-
[33] Choyke PL, Austin HA, Frank JA, Girton ME, Diggs parison with 99Tc-MAG3 dynamic scintigraphy. Magn
RL, Dwyer AJ, et al. Hydrated clearance of gado- Reson Imaging 1994;12:413 – 9.
linium-DTPA as a measurement of glomerular filtra- [45] Prasad PV, Goldfarb J, Sundaram C, Priatna A, Li W,
tion rate. Kidney Int 1992;41:1595 – 8. Edelman RR. Captopril MR renography in a swine
[34] Dumoulin CL, Buonocore MH, Opsahl LR, Katzberg model: toward a comprehensive evaluation of renal
RW, Darrow RD, Morris TW, et al. Noninvasive mea- arterial stenosis. Radiology 2000;217:813 – 8.
surement of renal hemodynamic functions using gado- [46] Sharma RK, Gupta RK, Kumar A, Gupta A, Bhandari
linium enhanced magnetic resonance imaging. Magn M. Correlation of captopril magnetic resonance reno-
Reson Med 1994;32:370 – 8. gram and perfusion changes with severity of renal
[35] Niendorf ER, Grist TM, Lee Jr FT, Brazy PC, Santyr allograft artery stenosis. Transplant Proc 2001;33:
GE, et al. Rapid in vivo measurement of single-kid- 3381 – 2.
ney extraction fraction and glomerular filtration rate [47] Gonzalez R, Chiou R. The diagnosis of upper urinary
with MR imaging. Radiology 1998;206:791 – 8. tract obstruction in children: comparison of diuresis
[36] Coulam CH, Lee JH, Wedding KL, Spielman DM, Pelc renography and pressure flow studies. J Urol 1985;133:
NJ, Kee ST, et al. Noninvasive measurement of extrac- 646 – 9.
tion fraction and single-kidney glomerular filtration rate [48] Rohrschneider WK, Becker K, Hoffend J, Clorius JH,
with MR imaging in swine with surgically created renal Darge K, Kooijman H, et al. Combined static-dynamic
arterial stenoses. Radiology 2002;223:76 – 82. MR urography for the simultaneous evaluation of mor-
[37] Laurent D, Poirier K, Wasvary J, Rudin M. Effect of phology and function in urinary tract obstruction. II.
essential hypertension on kidney function as measured Findings in experimentally induced ureteric stenosis.
in rat by dynamic MRI. Magn Reson Med 2002;47: Pediatr Radiol 2000;30:523 – 32.
127 – 34. [49] Verswijvel GA, Oyen RH, Van Poppel HP, Goethuys
A.J. Huang, V.S. Lee / Radiol Clin N Am 41 (2003) 1001–1017 1017

had L, Brkovic D, et al. Magnetic resonance imaging [54] Huber A, Heuck A, Scheidler J, Holzknecht N, Baur A,
in the assessment of urologic disease: an all-in-one Stangl M, et al. Contrast-enhanced MR angiography in
approach. Eur Radiol 2000;10:1614 – 9. patients after kidney transplantation. Eur Radiol 2001;
[50] Nolte-Ernsting CC, Bucker A, Adam GB, Neuerburg 11:2488 – 95.
JM, Jung P, Hunter DW, et al. Gadolinium-enhanced [55] Arrazola L, Sutherland DE, Sozen H, Hunter DW,
excretory MR urography after low-dose diuretic injec- Payne WD, Najarian JS, et al. May-Thurner syndrome
tion: comparison with conventional excretory urogra- in renal transplantation. Transplantation 2001;71:
phy. Radiology 1998;209:147 – 57. 698 – 702.
[51] Chan JH, Tsui EY, Luk SH, Fung SL, Cheung YK, [56] Agildere AM, Tarhan NC, Bozdagi G, Demirag A, Ni-
Chan MS, et al. MR diffusion-weighted imaging of ron EA, Haberal M. Correlation of quantitative dynamic
kidney: differentiation between hydronephrosis and magnetic resonance imaging findings with pathology
pyonephrosis. Clin Imaging 2001;25:110 – 3. results in renal transplants: a preliminary report. Trans-
[52] Neimatallah MA, Dong Q, Schoenberg SO, Cho KJ, plant Proc 1999;31:3312 – 6.
Prince MR. Magnetic resonance imaging in renal trans- [57] Dörsam J, Knopp MV, Carl S, Oesingmann N, Schad
plantation. J Magn Reson Imaging 1999;10:357 – 68. L, Brkovic D, et al. Ureteral complications after kid-
[53] Fang YC, Siegelman ES. Complications of renal trans- ney transplantation: evaluation with functional mag-
plantation: MR findings. J Comput Assist Tomogr netic resonance urography. Transplant Proc 1997;29:
2001;25:836 – 42. 132 – 5.
Radiol Clin N Am 41 (2003) 1019 – 1035

Imaging of renal trauma


J. Kevin Smith, PhD, MD*, Philip J. Kenney, MD
Department of Diagnostic Radiology, University of Alabama at Birmingham Health System, 619 South 19th Street,
Birmingham, AL 35233, USA

Trauma is nondiscriminatory and affects children, dalities used in the evaluation of the trauma patient,
adolescents, young adults, pregnant women, and the and be familiar with those injuries sustained in blunt
elderly. Trauma is the second leading cause of years abdominal trauma.
of life lost for all Americans and the leading cause of Current trends in trauma care are for less invasive
death and disability for youth and young adult procedures and more conservative management of
Americans. The financial cost of injuries in America many injuries, including renal injuries [2,4,5]. Better
is estimated at more than $224 billion each year [1]. resuscitation techniques, organization of dedicated
Despite advances in the technology of motor vehicle trauma centers, and faster response times are chang-
safety, motor vehicle collision remains the most ing the way trauma surgeons evaluate patients. Im-
common cause of blunt abdominal trauma in the aging of trauma patients can help to determine which
United States. Other less frequent sources of blunt patients can be managed conservatively and which
trauma to the abdomen include falls from a height, patients may require surgery, and to improve long-
assaults, bicycle accidents, and horseback riding term patient outcome.
injuries. Renal injury is the most frequent urologic
trauma and occurs in up to 8% to 10% of patients
with significant blunt or penetrating abdominal trau- Selection of patients to image
ma; up to 80% of renal injuries are caused by blunt
trauma, mostly motor vehicle accidents, and most Most (95%) significant renal injuries are associated
significant renal injuries are associated with other with hematuria, but hematuria may be absent, espe-
major organ injuries [2,3]. cially with renal vascular injuries and ureteropelvic
Care of the traumatized patient requires a multi- junction (UPJ) avulsion or ureteral injuries [6,7]. Only
disciplinary approach. The goal of trauma care is to about 1 to 5 out of 1000 blunt trauma patients with only
resuscitate the patient, to diagnose injuries, and to microscopic hematuria and without hypotension have
implement appropriate therapeutic measures as quick- significant urinary tract injury [8 – 11], so microhema-
ly as possible. Radiologists largely play a role in the turia alone is not an absolute indication for imaging.
diagnosis and staging of injuries. Interventional radi- At the authors’ institution abdominal and pelvic CT
ologists play an additional role in the management of is routinely used for blunt trauma patients with
arterial injuries using angiography with transcatheter abdominal symptoms, hypotension, or significantly
embolization. To be an effective member of the depressed level of consciousness. CT is used for
trauma team, the radiologist must be available for evaluation specifically of the genitourinary (GU) tract
emergent consultation, be adept at the imaging mo- for patients with gross hematuria; microscopic hema-
turia and hypotension; or patients with injuries asso-
ciated with renal injuries, such as lumbar spine, lower
* Corresponding author. N358 Jefferson Towers, De- rib, or transverse process fractures. Patients with pene-
partment of Radiology, University of Alabama Hospital, 619 trating trauma and any degree of hematuria undergo
South 19th Street, Birmingham, AL 35249 – 6830. urologic imaging. CT of all pediatric trauma patients
E-mail address: jksmith@uabmc.edu (J.K. Smith). with any hematuria, even microscopic hematuria

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00075-7
1020 J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035

alone, has traditionally been advocated but recent evi- cystography, and retrograde urethrography. With
dence suggests an approach similar to adult trauma pa- ready availability of CT the IVU has taken a more
tients may be acceptable [12]. limited role because of its lower sensitivity for in-
jury, lesser sensitivity for urinary contrast extravasa-
tion, and lack of ability to detect nonurologic injuries
Imaging modalities (Fig. 1) [13,14]. The IVU still may be used if CT
is not readily available, for unstable patients going to
Radiography surgery, or for urologic imaging if the patient is al-
ready in the operating room. This is typically per-
Radiography is an important tool in the primary formed as a one-shot intravenous pyelogram, which
evaluation of chest and skeletal trauma; however, its actually consists of a scout radiograph and typically
use in the setting of blunt abdominal trauma is one film immediately after contrast injection and
virtually nonexistent. Previously described signs of another about 10 minutes after contrast injection.
hemoperitoneum on radiography are not of sufficient Additional delayed films may be needed if there is
sensitivity or specificity to be useful. The widespread delayed excretion of contrast and to detect urinary
availability of CT, and to some degree ultrasonogra- contrast extravasation. The IVP may demonstrate loss
phy (US), has replaced abdominal radiography in this of the renal outline or psoas shadow if there is
regard. Radiography does still play a role in the perinephric hemorrhage, diminished or nonexcretion
setting of penetrating trauma to the abdomen. (Fig. 2), or contrast extravasation from an injured
kidney. The ureters should be visualized to evaluate
Intravenous urography for ureteral injury or displacement and contralateral
functioning kidney confirmed if there is significant
Traditionally, genitourinary injury has been renal injury, in the event the injured kidney may need
assessed by intravenous urography (IVU), standard to be removed [5,15].

Fig. 1. (A) Ten-minute radiograph from a one-shot intravenous pyelogram on a patient involved in a motor vehicle collision
shows normal kidneys and ureters bilaterally. (B) Image from contrast CT of the abdomen shows a splenic laceration with active
contrast extravasation (arrow).
J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035 1021

right upper quadrant including the hepatorenal recess,


(2) the left upper quadrant including the splenorenal
recess, (3 and 4) both paracolic gutters, (5) the pelvis
including its various peritoneal cavity recesses, and
(6) the pericardial space [19].
Various studies have proposed using US to search
for solid organ injury, but sufficient sensitivities and
specificities have not been demonstrated [20 – 22].
US has also been used to screen all blunt abdominal
trauma victims as part of a management algorithm. In
some institutions, radiologists or sonologists perform
the US examination, but in many centers this task
falls to the trauma surgeon or emergency physician.
There is little satisfactory training for clinical US in
these specialties and virtually no training in the
technical aspects of US; the ability of such individ-
uals to perform quality examinations has been seri-
ously questioned. If trauma US is to be performed by
the radiology department, the service must be readily
Fig. 2. Absent nephrogram. Ten-minute radiograph from a available at all times.
one-shot intravenous pyelogram of a patient involved in a Ultrasound may show renal laceration or a change
motor vehicle collision shows no enhancement or contrast in echogenicity of the injured kidney, or a decrease in
excretion on the right. the usual perinephric echogenicity if there is peri-
nephric fluid or hemorrhage. If US is negative and
Ultrasound there is significant hematuria, or if the US is positive,
CT is still indicated for better evaluation of the injury
The use of abdominal US in trauma patients if the patient is stable. For this reason the use of US is
continues to be controversial; the use of US for probably best reserved for rapid evaluation for intra-
detection of renal and urologic injuries is particularly peritoneal fluid in the unstable patient who may
problematic. US is able to detect free fluid in the require urgent surgery.
abdomen and pelvis but it cannot distinguish between
extravasated urine, blood, and other types of fluid, an Angiography
often clinically important distinction, and cannot
determine the source of bleeding. US is less sensitive Before the widespread availability of CT, angiog-
at depicting solid organ injury, especially of the raphy was often used to evaluate renal abnormalities
kidneys, depicting as few as 22% of renal injuries seen at IVU, especially suspected arterial injuries.
[16,17]. Although significant renal injuries are often With the advent of faster CT scanners and their
associated with other abdominal injuries, isolated increased detection of active arterial extravasation,
renal injuries may not have associated peritoneal fluid angiography is being used less frequently for the
in as many as 65% of the cases (Fig. 3) [16]. In initial diagnosis of traumatic injuries. CT shows
addition, US is insensitive for retroperitoneal blood many injuries not seen at angiography and accurately
and hollow organ injury [18]. characterizes most vascular injuries. Even vascular
Nevertheless, US has gained moderate acceptance contrast extravasation is better depicted by CT. Con-
in the United States as a means to evaluate the patient versely, the role of angiography in the management of
with blunt abdominal trauma. US in the setting of vascular and exsanguinating solid-organ injuries con-
trauma usually consists of a focused abdominal tinues to increase given the emphasis on nonoperative
sonography for trauma (FAST) scan. FAST scans management of trauma patients. Angiography with
can be completed in several minutes during the transcatheter embolization is becoming the standard
resuscitation of the patient in the trauma bay. The of care in the treatment of patients with many
primary goal of the FAST scan is the identification of vascular injuries. Angiographic embolization is well-
free fluid (hemoperitoneum) in the unstable patient, a suited to treat traumatic pseudoaneurysms and active
finding that usually prompts an exploratory laparot- arterial bleeding caused by splenic or hepatic and
omy. The FAST scan usually consists of interrogation sometimes renal injury, and hemorrhage associated
of six locations for the presence of free fluid: (1) the with pelvic ring injury [23 – 25].
1022 J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035

Fig. 3. Trauma ultrasound. (A) Ultrasound gray-scale image of a patient involved in a motor vehicle collision shows normal
right kidney. (B) Ultrasound image with power Doppler shows no blood flow within the right kidney. (C) Contrast-enhanced CT
image shows nonenhancing right kidney. Note relatively small amount of hemorrhage and the blind ending stump of the renal
artery (arrow).

Diagnostic peritoneal lavage lavage is not specific for the type of injury. Some
authors argue screening diagnostic peritoneal lavage
The diagnostic peritoneal lavage is not an imag- with selective use of CT is less expensive, but actual
ing modality but is the traditional gold standard for cost analysis is lacking [31]. At the authors’ institu-
evaluating for abdominal injury, and is safe and tion, there is a multidetector CT scanner in the emer-
rapid when performed by experienced surgeons. gency department within a few feet of the trauma
Early studies showed diagnostic peritoneal lavage bay and diagnostic peritoneal lavage is now infre-
to be faster and more rapid than CT, and many quently performed.
showed diagnostic peritoneal lavage to have better
accuracy [26 – 29]. With improvements in CT tech- Retrograde pyelography
niques including dynamic and then helical scanning,
increased experience with CT, and location of CT in Retrograde pyelography is primarily useful if
close physical proximity to the trauma bay, however, ureteral, UPJ, or renal pelvic injury is suspected
these differences have been erased [30]. For renal and delayed images were not obtained or were not
injuries diagnostic peritoneal lavage, like ultrasound, adequate to exclude these injuries on CT or IVU. It
may be especially problematic because isolated renal is often not practical in the emergent evaluation of
injuries may not be associated with intraperitoneal the severely injured patient, however, and does not
fluid, and when positive the diagnostic peritoneal characterize renal parenchymal injuries.
J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035 1023

Radionuclide renal scintigraphy examinations can be performed without having to


wait for the CT tube to cool.
Radionuclide renal scintigraphy may be used to Intravenous contrast is a necessity for satisfactory
evaluate the renal function of injured kidneys, eval- accuracy in abdominal CT scans performed for trau-
uate perfusion of a kidney with demonstrated or ma. Solid organ injuries, such as liver, splenic, or
suspected arterial injury, and for evaluation of the renal lacerations, can be unapparent on noncontrast
repaired kidney or renal vasculature. Since the wide- scans. Active arterial extravasation can only be
spread availability of CT, renal scintigraphy is rarely detected with the use of intravenous contrast. Low-
used in the acute setting. osmolality, nonionic contrast is preferred. A typical
contrast dose is 120 to 150 mL for adults and 1.5 to
MR imaging 2 mL/kg for children. An injection rate of at least
2 mL/second is desirable, but rates in the range of 3 to
MR imaging with gadolinium may be helpful to 4 mL/second provide optimal vascular and parenchy-
assess or characterize renal injury in the stable patient mal enhancement. Helical CT and multidetector
with strong contraindication for iodinated contrast, CT scanners have increased the frequency with which
but MR imaging is usually not practical in the acutely active arterial extravasation can be detected.
severely injured patient because of motion artifacts Most authors favor the use of oral contrast in
and the time often required. trauma abdominal CT scans. Oral contrast is safe,
even in children [32,33]. Administration of oral
CT contrast can aid greatly in the detection of bowel
injuries. A dilute solution of 4% diatrizoate meglu-
CT is the most comprehensive diagnostic tool mine in tap water is administered by mouth or by
available for the evaluation of the victim of blunt nasogastric tube as soon as the abdominal CT is
abdominal trauma. Unlike US and diagnostic perito- requested. A volume of 400 to 600 mL is given.
neal lavage, which are limited to answering certain For trauma patients the scan is not delayed for
specific diagnostic questions (eg, is there hemo- passage of oral contrast through the bowel. In this
peritoneum), CT affords a comprehensive evaluation short time frame, usually only the stomach, duode-
of all the intra-abdominal structures. One of the major num, and proximal jejunum are opacified. Fortunate-
advantages of CT is its ability to stage injuries to the ly, these are some of the most common sites of bowel
abdomen. The trend toward greater nonoperative injury. Some authors suggest withdrawing the naso-
management of traumatic abdominal injuries can be gastric tube into the distal esophagus during the scan
attributed in large part to successful staging of inju- to reduce streak artifact in the upper abdomen.
ries by CT. It is desirable to have the CT scanner as Five millimeters or less image thickness is helpful
close to the trauma bay as possible to minimize pa- to avoid significant volume averaging artifacts. For
tient transport time. single-slice helical CT a scanner pitch of 1.5:1 is a
good compromise between speed and excessive slice
CT technique profile broadening. On a multidetector CT scanner
Optimal evaluation of the blunt abdominal trauma high-speed (pitch greater than one) scanning speeds
victim requires optimization of CT technique. Ade- the image acquisition and still generally results in
quate scans can be obtained on conventional axial CT excellent image quality. It may be helpful to scan at
scanners, but helical CT scanners offer a substantial less than the maximum table speed to allow retro-
gain in speed and quality. Multidetector CT scanners spective reconstruction of thinner slices if needed for
have given the trauma radiologist an even more subtle injuries or the evaluation of associated spine or
powerful tool compared with single-slice helical CT. bony pelvic injuries. For example, on a General
Thin-section, high-quality images can be obtained in Electric four-slice scanner the authors use HS mode
a fraction of the time required for even helical single- with 5-mm images and table speed of 15 mm per
slice scanners. Shorter scan times mean less time for rotation, 0.8-second scanning. This allows retrospec-
motion and breathing artifact. Multidetector CT tive reconstruction of 2.5-mm thick slices if needed.
allows for optimal detection of injuries, such as active With the newer 16 or more slice scanners even the
arterial contrast extravasation, while decreasing the fastest table speeds still allow reconstruction of very
time that critically injured trauma patients are re- thin slices if needed. Kilovolt (peak) is usually 140
quired to be in the CT scanner. Multidetector CT is and milliampere seconds between 100 and 300 de-
not only faster but also offers much more efficient use pending on scan mode and patient size. The acquisi-
of tube-heat capacity so that multiple, consecutive CT tion start times after beginning contrast injections are
1024 J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035

Fig. 6. Perinephric and subcapsular hematomas. Contrast-


enhanced CT scan of a patient involved in a motor vehicle
collision shows an ill-defined high-density fluid collection
in the perinephric space (arrows). This patient also had a
subcapsular hematoma with deformity of the renal paren-
Fig. 4. Renal contusion. Contrast-enhanced CT of a patient chyma (arrowheads).
in a motor vehicle collision with small, ill-defined wedge-
shaped area of slight hypoenhancement in the mid right they are obtained while the patient is still on the CT
kidney (arrow).
scanner and if there is significant perinephric or peri-
ureteral fluid, delayed images (10 to 15 minutes) are
45 seconds for chest and 75 seconds for abdomen; a obtained to evaluate for urinary contrast extravasation
pause of 180 seconds before scanning the pelvis al- Standard CT with intravenous contrast has been
lows the bladder to opacify if a CT cystogram is not shown to yield up to 40% false-negatives for bladder
going to be performed. Some centers routinely scan injury. If bladder injury is a clinical concern (gross
through the kidneys a second time during the uro- hematuria or pelvic ring fracture), a cystogram or CT
graphic phase of enhancement to detect subtle paren- cystogram should be performed. After the initial scan,
chymal and collecting system injuries. The authors’ the patient can be evaluated by CT cystography
trauma patient’s images are routinely evaluated as without having to move to another location. CT cys-
tography is equal to or better than conventional cys-
tography if adequate retrograde bladder distention is
achieved with dilute contrast and CT cystography is
capable of distinguishing intraperitoneal, extraperito-

Fig. 5. Subcapsular hematoma. Contrast-enhanced CT scan of


a patient involved in a motor vehicle collision demonstrates a Fig. 7. Grade 2 – 3 renal injury: renal laceration. Contrast-
crescentic high-density fluid collection around the left kidney enhanced CT scan of a patient involved in a motor vehicle
(arrows). Note the relatively well-defined outer margin and collision shows slightly irregular low-attenuation defect in
the deformity of the underlying renal parenchyma. the anterior left kidney with associated perinephric fluid.
J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035 1025

Fig. 8. Grade 3 renal injury: renal laceration. (A) Contrast-enhanced portal venous phase image from a CT scan of a patient involved
in a motor vehicle collision has an irregular nonenhancing renal parenchymal defect in the mid lateral left kidney with associated
perinephric hematoma. (B) Delayed image from the same CT scan shows no urinary contrast extravasation.

neal, or combined bladder rupture [34]. For CT or high-riding prostate gland on physical examination
cystography, once the abdominal CT scan is com- should undergo a retrograde urethrogram before
plete, the urinary bladder is drained by Foley catheter. placement of a Foley catheter. Patients with a lower
The bladder is then filled by gravity infusion with index of suspicion can undergo a pericatheter urethro-
dilute iodine-based contrast (12 mL of 300-strength gram at a later time and a catheter is generally placed
contrast in 500 mL normal saline) and the pelvis during the initial assessment in the trauma bay.
scanned again when the bladder is fully distended
taking care to include the entire urinary bladder. CT interpretation
Trauma patients with histories consistent with Thoroughness and attention to detail are of vital
urethral trauma, gross blood at the urethral meatus, importance in the interpretation of CT scans for blunt

Fig. 9. Grade 4 renal injury: renal laceration into collecting system. (A) Tiny amount of fluid medial to the renal pelvis (arrow)
was the only clue to this laceration involving the collecting system. (B) Delayed image from the same CT shows a tiny area of
urinary contrast extravasation (arrow).
1026 J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035

Fig. 10. Grade 4 renal injury: lacerations extending into the collecting system. (A) Contrast-enhanced CT scan of a patient
involved in a motor vehicle collision shows several deep lacerations into the collecting system (arrows). (B) Delayed image from
the same CT scan shows urinary contrast extravasation (arrow).

abdominal trauma. A complete evaluation of a trauma and gluteals; bones including the spine and pelvis;
CT involves viewing the entire scan with three dif- and thighs (looking for soft tissue hematoma). West
ferent window-level settings: (1) soft tissue, (2) lung, [35] describes this systematic review as the ‘‘every-
and (3) bone. Immediate life-threatening injuries organ-on-every-slice’’ approach. The authors believe
should be sought first: large hemoperitoneum, large image review on the modern PACs workstation is
or tension pneumothorax, pneumoperitoneum, signs best done by paging relatively rapidly through the
of hypovolemic shock, and active arterial extravasa- images multiple times, paying specific attention to a
tion. Each area of the abdomen and pelvis should specific organ during each pass through the images.
then be interrogated for the presence of injury: liver The authors prefer to describe this approach for
and right paracolic gutter; spleen and left paracolic image review as ‘‘every-slice-of-every-organ.’’
gutter; upper abdominal organs including the stom-
ach, duodenum, pancreas, gallbladder, and biliary
tree; retroperitoneum including the adrenals, kidneys, Classification of renal injuries
inferior vena cava, and aorta; small bowel, colon, and
mesentery; pelvis including the urinary bladder; Renal injuries are graded by the American Asso-
muscles including the abdominal wall, psoas, iliacus, ciation for the Surgery of Trauma according to the

Fig. 11. Grade 4 renal injury: segmental infarctions. (A) Contrast-enhanced CT scan of a patient involved in a motor vehicle
collision shows well-defined, wedge-shaped, nonenhancing areas in the mid left kidney. (B) Follow-up contrast-enhanced CT scan
of the same patient shows complete resolution of the findings in about 2 weeks.
J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035 1027

Contusions
Nonexpanding subcapsular hematomas
Grade 2
Nonexpanding perinephric hematomas con-
fined to the retroperitoneum
Superficial cortical lacerations less than 1 cm
in depth without collecting system injury
Grade 3
Renal lacerations greater than 1 cm in depth
that do not involve the collecting system
Grade 4
Renal lacerations extending through the kid-
ney into the collecting system
Injuries involving the main renal artery or vein
with contained hemorrhage
Segmental infarctions without associated lac-
erations
Grade 5
Fig. 12. Grade 4 renal injury: infarctions and associated
Shattered or devascularized kidney
laceration. Contrast-enhanced CT scan of a patient involved UPJ avulsions
in a motor vehicle collision shows well-circumscribed, Complete laceration or thrombus of the main
wedge-shaped areas of nonenhancement posteromedially in renal artery or vein
both kidneys (arrows). The right kidney has associated
laceration and hematoma. Note also the peritoneal fluid and These scores were devised principally to facilitate
bowel edema (shock bowel). clinical research, but the radiologist should be familiar
with the scoring system because it is part of the
depth of the injury and involvement of vessels or the language of evaluation and triage use by the trauma
collecting system as follows[36]: surgeon. In general the American Association for
the Surgery of Trauma injury grade correlates with
Grade 1 the perceived need for surgery to repair or remove the
Hematuria with normal imaging studies injured kidney [37]. Even with high-grade injuries,

Fig. 13. Grade 5 renal injury: infarctions and multiple deep lacerations. (A) Contrast-enhanced CT scan of a patient involved in a
motor vehicle collision shows a well-defined, wedge-shaped, nonenhancing infarction (arrow) and several deep, irregular,
nonenhancing lacerations. (B) Delayed image from the same CT scan has contrast extravasating medially and laterally from the
deep lacerations.
1028 J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035

however, nonoperative management may be success- Grade 1 injuries


ful or even preferred in stable patients because renal
function is often better preserved with nonoperative American Association for the Surgery of Trauma
management [5,38]. Unless there is extensive devital- grade 1 renal injuries account for about 80% of renal
ized tissue, active hemorrhage, a large injury to the injuries and include hematuria with normal imaging
collecting system, or ureteral disruption, renal injuries studies, contusions, and nonexpanding subcapsular
are often managed conservatively [39,40]. hematomas. Contusions are seen as either ill-defined

Fig. 14. Grade 5 renal injuries: shattered kidney with venous injury managed conservatively. (A) Contrast-enhanced CT scan of a
patient involved in a motor vehicle collision demonstrates nearly occlusive thrombus in the right renal vein (arrow). (B) A slightly
lower image from the same CT scan has multiple deep, irregular hypodense lacerations extending through the renal parenchyma
with hematoma within the lacerations and around the kidney and devitalized segments of renal parenchyma (arrow). (C) Delayed
image from the same CT scan shows faint residual area of vascular contrast extravasation (arrow), which is much less dense than the
adjacent ureteral contrast. (D) Follow-up contrast-enhanced CT scan on the same patient. The kidney is deformed but the renal vein
injury and the urinary leak resolved and there is considerable residual functioning renal parenchyma.
J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035 1029

or sometimes sharply marginated areas of decreased clot, and may be higher than water density but are
enhancement and excretion. They are distinguishable without enhancement. Grade 2 lacerations are defined
from segmental infarctions by the presence of en- as less than 1 cm in depth and without involvement of
hancement (Fig. 4). Subcapsular hematomas are less the collecting system (Fig. 7), and have no urinary
common in blunt trauma than perinephric hematomas contrast extravasation. Grade 3 renal injuries include
and show up as an often high-density fluid collection similar renal lacerations that are greater than 1 cm,
contained between the renal parenchyma and the but do not involve the collecting system (Fig. 8).
renal capsule, frequently with some deformity of Grade 1 to 3 renal injuries are almost always man-
the underlying kidney. When small they may be aged conservatively unless there is brisk active hem-
crescentic (Fig. 5), but larger collections may become orrhage [39,40]. Active hemorrhage may be managed
elliptical and compress the renal parenchyma (Fig. 6). successfully with selective catheter embolization if
Rarely, the collection may compress the kidney the patient is otherwise stable [41,42].
enough to decrease the renal perfusion and result in
reactive hypertension, the so-called ‘‘Page kidney.’’
Grade 4 injuries
Grade 2 and 3 injuries
Renal lacerations extending through the kidney
Grade 2 injuries include nonexpanding perinephric into the collecting system or injuries involving the
hematomas confined to the retroperitoneum and main renal artery or vein with contained hemorrhage
superficial cortical lacerations less than 1 cm in depth are classified as grade 4. Lacerations involving the
without collecting system injury. The perinephric collecting system frequently lead to extravasation of
hematoma may be an isolated injury but is often urine and urinary contrast; any time there are lacer-
associated with underlying renal injury. It manifests ations extending through the kidney or significant
on CT as a typically ill-defined, often high-density perinephric fluid, especially around the renal hilum,
fluid collection between the renal parenchyma and delayed images should be obtained to evaluate for
Gerota’s fascia (see Fig. 6). The presence of a peri- urine extravasation (Figs. 9, 10). Even large urinary
nephric hematoma should prompt a thorough search extravasations often resolve with conservative treat-
for an underlying renal injury. A perinephric hema- ment, but stenting may be helpful with larger leaks. If
toma may be quite large, but traditionally does not there is significant devitalized renal tissue, especially
deform the kidney as opposed to the typical sub- with concomitant intraperitoneal injuries, in addition
capsular hematoma, which often does deform the renal to the urine leak, surgical debridement or repair may
contour when large. be needed to prevent later development of urinoma
Renal lacerations appear as irregular or linear and infection or abscess formation, which may ne-
parenchymal defects, which may contain blood or cessitate nephrectomy to prevent sepsis [4].

Fig. 15. Grade 5 renal injury: ureteropelvic junction avulsion. (A) Five-minute film from an intravenous pyelogram on a patient
involved in a motor vehicle collision shows relatively minor calyceal blunting on the left. (B) Ten-minute film shows progressive
accumulation of urinary contrast adjacent to the blind ending proximal ureter. The lack of contrast in the more distal ureter
suggests a complete tear.
1030 J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035

Fig. 16. Grade 5 renal injury: ureteropelvic junction avulsion. (A) Contrast-enhanced CT of a patient involved in a motor vehicle
collision has a small amount of medial perinephric fluid (arrows). (B) Delayed image from the same CT scan shows medial
perinephric urinary contrast extravasation and no contrast in the more distal ureter.

Segmental infarctions without associated lacera- tension may develop as a delayed complication in
tions are also grade 4 injuries. Segmental infarctions 6% to 20% of patients but often resolves or can be
may occur because of thrombosis, dissection, or lac- medically managed [25,45].
eration of segmental arteries and are often associated
with other renal injuries. They manifest as well- Grade 5 injuries
circumscribed, linear or wedge-shaped, often multi-
focal nonenhancing areas extending through the renal Injuries resulting in a shattered or devascularized
parenchyma in a radial or segmental orientation kidney, UPJ avulsions, and complete laceration or
(Figs. 11, 12). They usually resolve spontaneously thrombosis of the main renal artery or vein are
(see Fig. 11) or result in relatively minor renal scaring classified as grade 5 renal injuries. A shattered kidney
and are treated conservatively [43,44]. Rarely hyper- is basically the extreme of multiple renal lacerations,

Fig. 17. Grade 5 renal injury: ureteropelvic junction avulsion. (A) Contrast-enhanced CT of a patient involved in a motor vehicle
collision has a moderate perinephric fluid collection (straight arrows) and minimal medial perinephric fluid (white arrow).
(B) Delayed images of the same CT scan demonstrate urinary contrast extravasation and lack of opacification of the distal ureter.
J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035 1031

and there are often devitalized areas caused by fixed ureter and renal artery and vein. The UPJ injury
concomitant infarction, and urinary extravasation may be complete avulsion or partial tear. Both ex-
caused by associated injuries to the collecting system hibit characteristic medial or circumrenal urinoma
(Figs. 13, 14). (Figs. 15 – 17) [46 – 48]. The partial tear may be dis-
Injury of the UPJ occurs because of shearing tinguished from the complete avulsion by the pres-
stress at the renal pelvis; during rapid deceleration ence of contrast in the distal ureter [47]. Hematuria is
or hyperextension the kidney pulls on the relatively often absent [47,49]. Complete tears require surgical

Fig. 18. Grade 5 renal injury: missed ureteropelvic junction avulsion. (A) Contrast-enhanced CT on a patient after motor vehicle
collision shows a small amount of medial perinephric fluid (arrows). The significance of the finding was not appreciated, so
delayed images were not obtained. (B) Follow-up contrast-enhanced CT scan of the same patient shows interval development of
hydronephrosis and a large urinoma (arrows). (C) Percutaneous nephrostogram on the same patient shows interruption of the
ureter at or just below the ureteropelvic junction with extravasation of urinary contrast and no filling of the distal ureter. A
nephrectomy was eventually performed because of infection of the urinoma.
1032 J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035

repair, but some partial tears may resolve with stent-


ing or observation. If the diagnosis is missed and the
proximal collecting system is not drained, urinoma
may form and nephrectomy may be needed (Fig. 18).
A devascularized kidney most commonly results
from an incomplete vascular tear with thrombosis
involving the main renal artery. Hematuria is often
absent, especially if there are not other associated
injuries [50]. The kidney is nonenhancing, and there
may be little hematoma or other sign of injury (see
Fig. 3; Fig. 19). The blind ending renal artery is
sometimes seen and there may be retrograde opacifi-
cation of the renal vein or cortical rim sign, which may
not be apparent early [51]. More rarely, there is Fig. 20. Active vascular contrast extravasation. Contrast-
complete tear of the artery with massive hematoma enhanced CT on a patient in a motor vehicle collision with a
or active bleeding. These injuries are often associated tiny renal laceration, but with large subcapsular and peri-
with other renal injuries and this contributes to poor nephric hematomas and waterfall- shaped extravasation of
renal outcome of attempted repair so the management vascular contrast into the hematoma (arrow).
is usually expectant for stable patients or nephrectomy
if there is active bleed or major parenchymal disrup-
tion, unless there is injury or absence of the contralat- Vascular contrast extravasation
eral kidney [50,52]. Hypertension may develop as a
delayed complication weeks to months after the injury Bright enhancement close to the density of nearby
in as many as 40% to 50% of patients; often the arteries within a laceration or around an injured
hypertension resolves or can be managed medically kidney during the early phases of CT scanning
but occasionally nephrectomy is required [50,52]. indicates either contained or active hemorrhage. A
Injuries to the main renal vein are another less contained hemorrhage or pseudoaneurysm is fairly
common form of vascular pedicle injury. There may well circumscribed and contained within the renal
be thrombosis with CT typically showing filling
defect (see Fig. 14A) or nonenhancement of the vein
and delayed or persistent nephrogram with complete
occlusion [46,53]. Laceration of the renal vein
presents with medial or circumrenal subcapsular or
perinephric hematoma.

Fig. 21. Active vascular contrast extravasation. Contrast-


Fig. 19. Grade 5 renal injury: main renal artery. Contrast- enhanced CT on a patient in a motor vehicle collision with a
enhanced CT on a patient in a motor vehicle collision with small renal laceration, but with large subcapsular and peri-
nonenhancing right kidney. Note the relative lack of peri- nephric hematomas and flame-shaped extravasation of vas-
nephric fluid. There was no hematoma. cular contrast into the hematoma (arrow).
J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035 1033

parenchyma or laceration. Active hemorrhage is ill- renal trauma: a 7-year retrospective review from a pro-
defined or flame- or waterfall-shaped, with an asso- vincial trauma centre. Can J Urol 2001;8:1372 – 6.
ciated fresh hematoma, which often shows dependant [3] Sagalowsky AI, McConnell JD, Peters PC. Renal
trauma requiring surgery: an analysis of 185 cases.
or circumferential layering of older and fresher hem-
J Trauma 1983;23:128 – 31.
orrhage (Figs. 20, 21). As with other organs, active
[4] Moudouni SM, Patard JJ, Manunta A, Guiraud P,
extravasation of arterial contrast from the main renal Guille F, Lobel B. A conservative approach to major
artery or lacerated kidney may indicate the need for blunt renal lacerations with urinary extravasation and
urgent surgery or transcatheter embolization to pre- devitalized renal segments. BJU Int 2001;87:290 – 4.
vent exsanguination [54 – 58]. [5] Santucci RA, McAninch JM. Grade IV renal injuries:
Pseudoaneurysms may persist or enlarge and may evaluation, treatment, and outcome. World J Surg 2001;
occasionally cause delayed bleeding or rarely hyper- 25:1565 – 72.
tension. Arteriovenous fistulas associated with renal [6] Stables DP, Fouche RF, de Villiers van Niekerk JP, Cre-
lacerations from blunt or especially penetrating trau- min BJ, Holt SA, Peterson NE. Traumatic renal artery
occlusion: 21 cases. J Urol 1976;115:229 – 33.
ma may be unapparent initially but may enlarge over
[7] Boone TB, Gilling PJ, Husmann DA. Ureteropelvic
time, also potentially causing delayed bleeding, hy-
junction disruption following blunt abdominal trauma.
pertension, or high-output cardiac failure. J Urol 1993;150:33 – 6.
[8] Cass AS, Luxenberg M, Gleich P, Smith CS. Clinical
indications for radiographic evaluation of blunt renal
trauma. J Urol 1986;136:370 – 1.
Summary
[9] Nicolaisen GS, McAninch JW, Marshall GA, Bluth Jr
RF, Carroll PR. Renal trauma: re-evaluation of the in-
Trauma is a major cause of death and disability dications for radiographic assessment. J Urol 1985;133:
and renal injuries occur in up to 10% of patients 183 – 7.
with significant blunt abdominal trauma. Patients [10] Herschorn S, Radomski SB, Shoskes DA, Mahoney J,
with penetrating trauma and hematuria, blunt trau- Hirshberg E, Klotz L. Evaluation and treatment of
ma with shock and hematuria, or gross hematuria blunt renal trauma. J Urol 1991;146:274 – 6 [discus-
warrant imaging of the urinary tract specifically and sion: 6 – 7].
CT is the preferred modality. If there is significant [11] McAndrew JD, Corriere Jr JN. Radiographic evalua-
perinephric fluid, especially medially, or deep lacer- tion of renal trauma: evaluation of 1103 consecutive
patients. Br J Urol 1994;73:352 – 4.
ation, delayed images should be obtained to evaluate
[12] Perez-Brayfield MR, Gatti JM, Smith EA, Broecker B,
for urinary extravasation. Most renal injuries are Massad C, Scherz H, et al. Blunt traumatic hematuria
minor, including contusions, subcapsular and peri- in children. Is a simplified algorithm justified? J Urol
nephric hematoma, and superficial lacerations. More 2002;167:2543 – 6 [discussion: 6 – 7].
significant injuries include deep lacerations, shattered [13] McAninch JW, Federle MP. Evaluation of renal inju-
kidney, active hemorrhage, infarctions, and vascular ries with computerized tomography. J Urol 1982;128:
pedicle and UPJ injuries. These injuries are more 456 – 60.
likely to need surgery or have delayed complications [14] Federle MP, Kaiser JA, McAninch JW, Jeffrey RB,
but may still often be managed conservatively. The Mall JC. The role of computed tomography in renal
presence of urinary extravasation and large devi- trauma. Radiology 1981;141:455 – 60.
[15] Morey AF, McAninch JW, Tiller BK, Duckett CP, Car-
talized areas of renal parenchyma, especially with
roll PR. Single shot intraoperative excretory urography
associated injuries of intraperitoneal organs, is par- for the immediate evaluation of renal trauma. J Urol
ticularly prone to complication and usually requires 1999;161:1088 – 92.
surgery. Active hemorrhage should be recognized [16] McGahan JP, Richards JR, Jones CD, Gerscovich EO.
because it often indicates a need for urgent surgery Use of ultrasonography in the patient with acute
or embolization to prevent exsanguination. renal trauma. J Ultrasound Med 1999;18:207 – 13
[quiz: 15 – 6].
[17] McGahan JP, Rose J, Coates TL, Wisner DH, Newberry
P. Use of ultrasonography in the patient with acute ab-
References dominal trauma. J Ultrasound Med 1997;16: 653 – 62
[quiz: 63 – 4].
[1] National Center for Injury Prevention and Control. In- [18] Miller MT, Pasquale MD, Bromberg WJ, Wasser TE,
jury fact book 2001 – 2002. Atlanta (GA): National Cox J. Not so FAST. J Trauma 2003;54:52 – 9 [discus-
Center for Injury Prevention and Control, Centers for sion: 9 – 60].
Disease Control and Prevention; 2001. [19] McKenney KL, Nunez Jr DB, McKenney MG, Asher
[2] Baverstock R, Simons R, McLoughlin M. Severe blunt J, Zelnick K, Shipshak D. Sonography as the primary
1034 J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035

screening technique for blunt abdominal trauma: expe- Rue III LW. CT cystography: radiographic and clinical
rience with 899 patients. AJR Am J Roentgenol 1998; predictors of bladder rupture. AJR Am J Roentgenol
170:979 – 85. 2000;174:89 – 95.
[20] Bode PJ, Niezen RA, van Vugt AB, Schipper J. Ab- [35] West OC. Intraperitoneal abdominal injuries. In: West
dominal ultrasound as a reliable indicator for conclu- OC, Novelline RA, Wilson AJ, editors. Emergency and
sive laparotomy in blunt abdominal trauma. J Trauma trauma radiology: categorical course syllabus. Lees-
1993;34:27 – 31. burg (VA): American Roentgen Ray Society; 2000.
[21] Rothlin MA, Naf R, Amgwerd M, Candinas D, Frick p. 87 – 98.
T, Trentz O. Ultrasound in blunt abdominal and thora- [36] Moore EE, Shackford SR, Pachter HL, McAninch JW,
cic trauma. J Trauma 1993;34:488 – 95. Browner BD, Champion HR, et al. Organ injury scaling:
[22] Tso P, Rodriguez A, Cooper C, Militello P, Mirvis S, spleen, liver, and kidney. J Trauma 1989;29:1664 – 6.
Badellino MM, et al. Sonography in blunt abdominal [37] Santucci RA, McAninch JW, Safir M, Mario LA, Ser-
trauma: a preliminary progress report. J Trauma 1992; vice S, Segal MR. Validation of the American Associa-
33:39 – 43 [discussion: 4]. tion for the Surgery of Trauma organ injury severity
[23] Bretan Jr PN, McAninch JW, Federle MP, Jeffrey Jr scale for the kidney. J Trauma 2001;50:195 – 200.
RB. Computerized tomographic staging of renal trau- [38] Altman AL, Haas C, Dinchman KH, Spirnak JP. Se-
ma: 85 consecutive cases. J Urol 1986;136:561 – 5. lective nonoperative management of blunt grade 5 re-
[24] Hagiwara A, Sakaki S, Goto H, Takenega K, Fuku- nal injury. J Urol 2000;164:27 – 30 [discussion: 1].
shima H, Matuda H, et al. The role of interventional [39] Knudson MM, Maull KI. Nonoperative management
radiology in the management of blunt renal injury: of solid organ injuries: past, present, and future. Surg
a practical protocol. J Trauma 2001;51:526 – 31. Clin North Am 1999;79:1357 – 71.
[25] Bruce LM, Croce MA, Santaniello JM, Miller PR, [40] Brandes SB, McAninch JW. Reconstructive surgery for
Lyden SP, Fabian TC. Blunt renal artery injury: inci- trauma of the upper urinary tract. Urol Clin North Am
dence, diagnosis, and management. Am Surg 2001;67: 1999;26:183 – 99.
550 – 4 [discussion: 5 – 6]. [41] Corr P, Hacking G. Embolization in traumatic intra-
[26] Meyer DM, Thal ER, Weigelt JA, Redman HC. Eval- renal vascular injuries. Clin Radiol 1991;43:262 – 4.
uation of computed tomography and diagnostic perito- [42] Dinkel HP, Danuser H, Triller J. Blunt renal trauma:
neal lavage in blunt abdominal trauma. J Trauma 1989; minimally invasive management with microcatheter
29:1168 – 70 [discussion: 70 – 2]. embolization experience in nine patients. Radiology
[27] Kearney Jr PA, Vahey T, Burney RE, Glazer G. Com- 2002;223:723 – 30.
puted tomography and diagnostic peritoneal lavage in [43] Carroll PR, McAninch JW, Klosterman P, Greenblatt M.
blunt abdominal trauma: their combined role. Arch Renovascular trauma: risk assessment, surgical man-
Surg 1989;124:344 – 7. agement, and outcome. J Trauma 1990;30:547 – 52
[28] Fabian TC, Mangiante EC, White TJ, Patterson CR, [discussion: 53 – 4].
Boldreghini S, Britt LG. A prospective study of 91 pa- [44] Cass AS, Luxenberg M. Traumatic thrombosis of a seg-
tients undergoing both computed tomography and peri- mental branch of the renal artery. J Urol 1987;137:
toneal lavage following blunt abdominal trauma. 1115 – 6.
J Trauma 1986;26:602 – 8. [45] Bertini Jr JE, Flechner SM, Miller P, Ben-Menachem
[29] Marx JA, Moore EE, Jorden RC, Eule Jr J. Limitations Y, Fischer RP. The natural history of traumatic branch
of computed tomography in the evaluation of acute renal artery injury. J Urol 1986;135:228 – 30.
abdominal trauma: a prospective comparison with diag- [46] Harris AC, Zwirewich CV, Lyburn ID, Torreggiani
nostic peritoneal lavage. J Trauma 1985;25:933 – 7. WC, Marchinkow LO. CT findings in blunt renal trau-
[30] Pal JD, Victorino GP. Defining the role of computed ma. Radiographics 2001;21:S201 – 14.
tomography in blunt abdominal trauma: use in the [47] Kawashima A, Sandler CM, Corriere Jr JN, Rodgers
hemodynamically stable patient with a depressed level BM, Goldman SM. Ureteropelvic junction injuries sec-
of consciousness. Arch Surg 2002;137:1029 – 32 [dis- ondary to blunt abdominal trauma. Radiology 1997;
cussion: 32 – 3]. 205:487 – 92.
[31] Gonzalez RP, Ickler J, Gachassin P. Complementary [48] Kenney PJ, Panicek DM, Witanowski LS. Computed
roles of diagnostic peritoneal lavage and computed tomography of ureteral disruption. J Comput Assist
tomography in the evaluation of blunt abdominal trau- Tomogr 1987;11:480 – 4.
ma. J Trauma 2001;51:1128 – 34 [discussion: 34 – 6]. [49] Campbell Jr EW, Filderman PS, Jacobs SC. Ureteral
[32] Federle MP, Yagan N, Peitzman AB, Krugh J. Ab- injury due to blunt and penetrating trauma. Urology
dominal trauma: use of oral contrast material for CT 1992;40:216 – 20.
is safe. Radiology 1997;205:91 – 3. [50] Haas CA, Dinchman KH, Nasrallah PF, Spirnak JP.
[33] Lim-Dunham JE, Narra J, Benya EC, Donaldson JS. Traumatic renal artery occlusion: a 15-year review.
Aspiration after administration of oral contrast material J Trauma 1998;45:557 – 61.
in children undergoing abdominal CT for trauma. AJR [51] Kamel IR, Berkowitz JF. Assessment of the cortical
Am J Roentgenol 1997;169:1015 – 8. rim sign in posttraumatic renal infarction. J Comput
[34] Morgan DE, Nallamala LK, Kenney PJ, Mayo MS, Assist Tomogr 1996;20:803 – 6.
J.K. Smith, P.J. Kenney / Radiol Clin N Am 41 (2003) 1019–1035 1035

[52] Knudson MM, Harrison PB, Hoyt DB, Shatz DV, Ziet- dynamic contrast-enhanced CT. AJR Am J Roentgenol
low SP, Bergstein JM, et al. Outcome after major 1991;156:725 – 9.
renovascular injuries: a Western trauma association [56] Lane MJ, Katz DS, Shah RA, Rubin GD, Jeffrey Jr
multicenter report. J Trauma 2000;49:1116 – 22. RB. Active arterial contrast extravasation on helical CT
[53] Blankenship B, Earls JP, Talner LB. Renal vein throm- of the abdomen, pelvis, and chest. AJR Am J Roent-
bosis after vascular pedicle injury. AJR Am J Roent- genol 1998;171:679 – 85.
genol 1997;168:1574. [57] Shanmuganathan K, Mirvis SE, Sover ER. Value of
[54] Federle MP. CT of active hemorrhage from abdominal contrast-enhanced CT in detecting active hemorrhage
and pelvic trauma. In: West OC, Novelline RA, Wilson in patients with blunt abdominal or pelvic trauma. AJR
AJ, editors. Emergency and trauma radiology: catego- Am J Roentgenol 1993;161:65 – 9.
rical course syllabus. Leesburg (VA): American Roent- [58] Willmann JK, Roos JE, Platz A, Pfammatter T, Hilfiker
gen Ray Society; 2000. p. 79 – 86. PR, Marincek B, et al. Multidetector CT: detection of
[55] Jeffrey Jr RB, Cardoza JD, Olcott EW. Detection of active hemorrhage in patients with blunt abdominal
active intraabdominal arterial hemorrhage: value of trauma. AJR Am J Roentgenol 2002;179:437 – 44.
Radiol Clin N Am 41 (2003) 1037 – 1051

Imaging of hereditary renal cancer


Peter L. Choyke, MD
Department of Radiology, Imaging Sciences Program, National Institutes of Health, NIH Building 10, Room 1C660,
Bethesda, MD 20892 – 1182, USA

Renal cancer is diagnosed in over 30,000 Ameri- Histologic subtypes of renal cancer
cans each year and accounts for approximately
12,000 annual deaths. Smoking, obesity, and occu- Before considering the individual hereditary renal
pational exposures have been implicated in the cancer syndromes it is important to review the char-
development of renal cancers but, in general, the acteristics of the different cell types of renal cancer.
cause of renal cancer remains obscure [1]. Although Renal cancers can be subclassified into a variety of cell
hereditary renal cancer makes up only approximately types (Fig. 1) [5]. Clear cell carcinomas are the most
4% of the total number of cases, this number is frequent type of renal cancer accounting for approxi-
expected to grow as a more complete understanding mately 75% of renal cancers. The term ‘‘clear cell
of the genetic causes of cancer is elucidated [2]. As carcinoma’’ encompasses the clear cell variant, the
hereditary renal cancer syndromes become better granular cell variant, and mixed cell types. The high
understood, they provide insights into the mecha- glycogen content within the cytoplasm of clear cell
nisms of cancer development in the general popula- cancer cells accounts for their lucent appearance on
tion and assist efforts to prevent and treat renal conventional histologic stains. When glycogen is less
cancers (Table 1). abundant, the cytoplasm is darker and the cells are
The most common cell type of renal cancer is the termed ‘‘granular.’’ A delicate but rich and permeable
clear cell carcinoma, followed by papillary (types I vascular supply is often seen throughout these tumors,
and II), chromophobe carcinoma and oncocytoma although regions of necrosis, fibrosis, or hemorrhage
[3,4]. Medullary carcinoma and duct of Bellini can- are avascular or hypovascular. Clear cell carcinomas
cers are rare renal tumors. Over the past 5 years, are thought to arise from the proximal tubular epithe-
hereditary renal cancer syndromes have been asso- lium of the kidney
ciated with one or more of these cancer cell types. The second leading type of renal cancer is termed
The genes responsible for these syndromes have been ‘‘papillary,’’ also sometimes called ‘‘chromophil’’
discovered in many cases and research is now under- renal cancer, which accounts for 10% to 15% of
way to explain the molecular pathways leading to all renal cancers. There are two subtypes of papillary
tumor development. A more complete picture of the renal cell carcinomas, type I and type II, which are
mechanisms underlying the development of tumors of distinguished by tumor architecture and cellular
varying cell types is emerging. morphology. Both types share a common papillary
Over the past decade substantial progress has been structure: a fibrovascular core with tumor cells lining
made in the understanding of the genetic basis of the surface of each papilla (see Fig. 1) [5,6]. Type I,
cancer in humans. This article reviews the current or basophilic renal cancer, usually is considered
state-of-the-science of hereditary renal cancers with clinically low grade and has a favorable prognosis.
particular attention to their imaging features and This tumor is composed of fronds of fibrovascular
clinical management. papillary and tubular structures covered by cells with
scanty cytoplasm and small oval nuclei. Foamy
macrophages, which are thought to represent a host
E-mail address: pchoyke@nih.gov immune response, are often present within the inter-

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00068-X
1038
Table 1
The hereditary renal cancers in adults

P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051


Genes ‘‘gene name’’ Frequency of Predominant renal Other renal
Syndrome (gene Product) renal cancer (%) tumor cell type tumor cell types Associated abnormalities
von-Hippel Lindau 3p26 ‘‘VHL’’ (pVHL) 28 – 45 Clear cell Cysts CNS hemangioblastomas, retinal
angiomas, pancreatic cysts,
neuroendocrine tumors of pancreas,
pheochromocytoma
Tuberous sclerosis 9q34 ‘‘TSC1’’ (hamartin) 16p13 1–2 Clear cell Cysts, angiomyolipoma, CNS tubers, angiofibromas of skin,
‘‘TSC2’’ (tuberin) papillary, chromophobe, cardiac rhadomyomas
oncocytoma
Hereditary papillary 7q34 ‘‘c-MET’’ (HGF receptor) 19 Papillary type1 None
renal cancer
Hereditary leiomyoma 1q42-43 ‘‘FH’’ (fumarate hydratase) 15 – 30 Papillary type 2 None Cutaneous and uterine leiomyomas
renal cell carcinoma
Birt-Hogg-Dubè 17p11.2 ‘‘BHD’’ (folliculin) 8 – 15 Chromophobe Clear cell, Fibrofolliculomas, lung cysts,
oncocytic neoplasm papillary, oncocytoma pneumothoraces
Familial renal oncocytoma Unknown Unknown Oncocytoma None Renal dysfunction
Medullary carcinoma 11p Unknown Medullary carcinoma None Sickle cell trait
of the kidney
P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051 1039

stitium and psamma bodies also are frequently Von Hippel-Lindau disease
present. Despite the apparent vascularity on his-
tology, type I papillary renal tumors typically enhance Von Hippel-Lindau disease (VHL) is a multisys-
poorly on CT or during renal angiography. Type II tem autosomal-dominant hereditary disorder charac-
papillary tumors, or eosinophilic renal cancers, bear a terized by the formation of hemangioblastomas in
superficial resemblance to type I tumors in that the the spine and posterior fossa, retinal angiomas,
basic frond-like architecture is present but they may pheochromocytomas, pancreatic cysts, cystadenomas
not be related at a biologic level. Type II papillary and neuroendocrine tumors, epididymal and broad
renal cancers consist of papillae covered by large ligament cystadenomas, and renal cysts and tumors.
cells with abundant eosinophilic cytoplasm and large VHL gives rise to a variety of renal cystic lesions
nuclei with prominent nucleoli. Type II papillary ranging from pure cysts to mixed solid and cystic
tumors often are more clinically aggressive than type masses to purely solid clear cell carcinomas of the
I papillary tumors and can enhance more intensely. kidney. The VHL gene, found at 3p25, is considered
Papillary tumors also are thought to arise from the an important housekeeping or tumor suppressor gene
proximal tubular epithelium that in its normal state functions to prevent the
The chromophobe carcinoma, so-named because development of renal cancers. Mutations or inactiva-
of its lack of staining with typical histologic stains, tion of the VHL gene are found in over 60% of
such as hematoxylin and eosin, is the third most sporadic clear cell renal carcinomas indicating that it
common type of renal cancer accounting for about is one of the crucial genes in the development of clear
5% of renal tumors. Chromophobe carcinoma can be cell carcinoma of the kidney [14,15].
stained with Hale’s colloidal iron, which yields a A tumor suppressor gene normally functions to
homogeneous blue cytoplasmic stain [7]. In routine decrease the chance of developing cancer. When a
histologic sections the cytoplasm tends to condense tumor suppressor gene is mutated and the resulting
near the cell membrane producing a halo around the protein product is abnormal, however, the affected
nucleus. The cytoplasm is rich in mitochondria cells are at increased risk of malignancy. Tumor
much like the oncocytoma [8]. The oncocytoma development requires that both copies of the gene
itself is considered a benign renal neoplasm; how- become mutated or deleted. Unlike other diseases in
ever, this categorization has been called into which more than one genetic locus has been impli-
question by the resemblance of oncocytoma to chro- cated, VHL seems to be caused by mutations at a
mophobe carcinoma and by reports of ‘‘metasta- single gene locus, 3p25 (short arm of third chromo-
sizing’’ oncocytomas [9 – 11]. Oncocytomas are some). The gene was first discovered in 1993 by Latif
comprised of cells with abundant eosinophilic cyto- et al [16]. Subsequently it has been demonstrated that
plasm that are filled with mitochondria accounting the VHL gene codes for a protein, pVHL. One of the
for the brown color on gross pathology. When they normal roles of pVHL is to assist in the degradation of
are numerous the term ‘‘oncocytosis’’ is used and an intracellular growth factor known as hypoxia
can be seen in patients with chromophobe carcino- inducible factor (HIF) [17]. HIF is an important
mas [11]. Chromophobe carcinomas and oncocyto- regulator of metabolism and is dependent on the
mas are thought to arise from intercalated cells in oxygen tension within a cell. Normally, HIF is pro-
the distal tubules. duced when the cell is exposed to hypoxic conditions.
Collecting duct carcinoma includes the medullary Once normal oxygen tension is restored, HIF is
renal cancer associated with sickle cell trait and duct quickly degraded in a process mediated by pVHL.
of Bellini tumors. Medullary renal cancer is charac- For the pVHL protein to interact with HIF and
terized histologically by irregular channels lined by mediate its degradation, it must first bind other sig-
highly atypical epithelium that sometimes have a naling intermediaries known as elongin b and c and
hobnail appearance. The channels are found in an Cul2 [18]. These molecules normally bind to pVHL
inflamed desmoplastic stroma. [12,13]. Both medul- allowing pVHL to bind to HIF, allowing ubiquitina-
lary renal cancer and duct of Bellini tumors are tion (ie, degradation) of HIF. The defective pVHL
clinically aggressive neoplasms. Medullary renal can- protein is unable to bind to these smaller molecules or
cer and its variants arise from the collecting ducts, binds only weakly. As a consequence HIF is not
which are histologically and embryologically distinct degraded even in normoxic conditions. The cell acts
from the tubular epithelium. as if it were chronically hypoxic even under normoxic
In the following sections the relationship between conditions. HIF also mediates the production of a
each of the preceding cell types of renal cancer and its number of downstream growth factors. These include
corresponding genetic syndrome are described. vascular endothelial growth factor, which is one of the
1040 P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051

Fig. 1. The cell types of renal cancer. (A) Clear cell carcinoma. (B) Type I papillary renal cancer. (C) Type II papillary renal
cancer. (D) Chromophobe carcinoma. (E) Oncocytoma.

critical growth factors in early tumor angiogenesis. from peripheral white cells is used to check for
Vascular endothelial growth factor has been a target recorded mutations in VHL. The test is 99% accurate
molecule for the first generation of antiangiogenic for the diagnosis of VHL [22].
treatments. One of the mechanisms of tumor devel- Renal lesions are a common manifestation of
opment in VHL may be the failure of pVHL to VHL. Between 60% and 70% of patients develop
suppress HIF leading to unregulated production of cysts in the kidney and about 40% develop radio-
vascular endothelial growth factor and angiogenesis. logically evident renal cancers (Fig. 2). If one exam-
[14,19]. Unregulated vascular endothelial growth fac- ines the normal-appearing tissue of a kidney from a
tor may account for the highly vascular nature of clear patient with VHL one finds hundreds of small tumor-
cell tumors of the kidney. HIF also stimulates the lets scattered throughout the parenchyma, invisible to
production of erythropoietin, glucose transporters the naked eye (Fig. 3) [23]. That only a few of many
(GLUT1), and nitric oxygen synthase. This could also tumors grow to visible tumors on CT is one of the
explain why some tumors in VHL produce excess unusual features of VHL. Tumors can take many
erythropoietin. Interestingly, when antibodies to vas- forms in VHL. They can appear as cysts, cystic renal
cular endothelial growth factor were administered to cancers, and solid renal cancers [24]. The cell lining
patients with VHL in a clinical trial, erythrocytosis of even a simple renal cyst in VHL contains clear
secondary to erythropoietin overproduction was cells similar in appearance to those found within clear
observed [20,21]. By blocking one pathway (vascular cell carcinomas.
endothelial growth factor), another (erythropoietin) Cysts can regress or grow. The solid components
was overexpressed. of mixed lesions tend to grow over time, whereas the
A variety of mutations of the VHL gene have been cystic areas remain unchanged or regress. Solid
discovered including complete and partial deletions, lesions tend to grow progressively [25]. Occasionally,
single missense mutations, and frame shift mutations. heavily calcified or hemorrhagic cystic disease is
Fortunately, patients suspected of the disease can now seen. Both are usually associated with less aggressive
be tested for VHL with a simple blood sample. DNA forms of renal cancer but this is not universally true.
P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051 1041

Renal cancers in VHL tend to be well differen-


tiated or Furman grade I to II when they are under
3 cm in diameter. Only one report of a metastatic
renal tumor less than 3 cm in diameter (2.5 cm) has
appeared in the literature [26]. As a consequence, it is
generally believed that treatment should be reserved
for patients with larger lesions, greater than or equal
to 3 cm in diameter. This strategy has been very
successful in preserving renal function while avoid-
ing metastatic disease [27,28].
Renal cancers are typically detected and measured
on serial contrast-enhanced CT scans (see Fig. 2).
Using multidetector CT, unenhanced images through

Fig. 3. Clear cell tumorlet in the parenchyma of a patient


with von Hippel-Lindau disease. Note that there is a tiny
focus of tumor within the renal parenchyma (arrow).
Hundreds of these lesions are found in the kidneys of
patients with VHL.

the liver and kidneys are first obtained with 5-mm


collimation. A bolus of intravenous iodinated contrast
(130 mL of a nonionic contrast agent) can be adminis-
tered at 3 mL/second and 2.5-mm thick sections
(reconstructed at 5-mm intervals) are obtained during
the arterial phase (approximately 25 seconds) and
during the venous phase (approximately 80 seconds).
The precontrast scans are useful for judging whether
a lesion is actually enhancing and are of particular use
when a hemorrhagic cyst is present. Arterial phase
images are useful for detecting pancreatic neuro-
endocrine tumors (found in approximately 10% to
15% of VHL patients), which enhance intensely only
on the arterial phase, whereas the venous phase is the
most important phase for evaluating the kidneys. The
adrenals are equally well seen on all three phases.
Three-dimensional CT angiography and reconstruc-
tion can also be performed as clinically necessary
before surgery where a three-dimensional model of
the kidney can be made to assist the surgeon in
identifying lesions for removal.
Serial CT imaging is recommended for patients
with VHL even if they have minimal or no renal
Fig. 2. (A, B) Von Hippel-Lindau disease. This 32-year-old disease. The lifetime risk of renal cancer is high in
man has bilateral solid and cystic renal masses. The solid VHL and the key to preserving renal function and
lesions proved to be clear cell carcinomas. preventing metastatic disease is careful monitoring of
1042 P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051

the patient [29]. Patients with minimal disease can be minimally invasive methods for treating small renal
scanned at 1- to 2-year intervals, whereas patients cancers in VHL and other hereditary renal masses
with active renal tumors need to be seen more fre- [33 – 35]. For lesions positioned close to the bowel,
quently (every 6 to 12 months). In patients with laparoscopically guided radiofrequency ablation or
lesions of borderline size (approximately 3 cm) even cryotherapy can be performed (Fig. 5) [36 – 38].
more frequent studies may be performed. Tumor Although no studies documenting improved survival
growth is not always linear and predictable and for or enhanced renal function have been reported, it
this reason the record of past CTs is not necessarily seems reasonable that these approaches result in less
predictive of the future for any given lesion. CT is the damage to the kidney. Because the tumor is not
preferred method of screening because it is relatively treated under direct vision, however, it also is
less expensive than MR imaging and the appearance possible that the tumor may be incompletely treated.
of renal masses is well understood. MR imaging with For this reason close follow-up with imaging studies
gadolinium chelate enhancement is a viable alterna- of treated lesions is important after radiofrequency
tive in patients with poor renal function, those wishing ablation or cryotherapy [37,38]. It is important to
to avoid ionizing radiation, or severe allergy to iodin- realize that an untreated remnant of tumor measur-
ated contrast media. This should be performed with a ing 5 mm in diameter from a tumor that was
torso array coil and fat suppression. Attempts should originally 3 cm behaves biologically like a 3-cm
be made to measure the lesion before and after lesion and not a 5-mm tumor. As a consequence,
contrast using the same parameters [30]. Ultrasound early retreatment of recurrences is recommended.
is less accurate than other techniques for detecting and Although radiofrequency ablation and cryotherapy
characterizing renal masses in VHL and should not be are attractive alternatives, long-term experience is
relied on exclusively [31]. still lacking.
Why wait to treat known tumors within the
kidney in a patient with VHL? This is a conscious
strategy based on the knowledge that VHL is a Hereditary papillary renal carcinoma
lifelong disorder that must be managed differently
than sporadic disease [28]. Preservation of renal Hereditary papillary renal cancer (HPRC) is an
function must be balanced against the risk of meta- autosomal-dominant hereditary condition in which
static renal cancer. Partial nephrectomy is the method the kidneys develop multiple, bilateral type I papil-
with the longest track record. Unfortunately, with lary renal cancers. No other extrarenal manifestations
every partial nephrectomy there is inevitably some have been reported in this syndrome. Sporadic type I
loss of renal function and scarring, which makes papillary renal tumors have a better prognosis than
subsequent surgeries even more difficult. As a con- other cell types, so it is not surprising that the tumors
sequence, surgeries should be spaced as far apart associated with HPRC tend to be slow growing and
from each other as possible in the hopes of pre- rarely cause death [39]. Because of the favorable
serving renal function for the longest possible inter- prognosis of HPRC, the patient may not come to
val. It is recommended that partial nephrectomies not medical attention and the disease often is not diag-
be performed until the solid component of the largest nosed until the patient is in their fifth decade. In
tumor is 3 cm or more. VHL, the patient is often diagnosed in their teens
The standard method of treatment of renal tumors and twenties.
in VHL is nephron-sparing surgery [27]. In this The gene responsible for HPRC has been located
procedure, the surgeon enucleates visible tumors at 7q 31.3 and is known as the c-MET proto-onco-
and cysts on the renal surface. Deeper lesions are gene [40]. This gene was first described in 1984 but
detected using intraoperative ultrasound (Fig. 4) [32]. was only recently linked to renal cancer [41]. Unlike
The purpose of the nephron-sparing approach is VHL where it is thought that a mutation leads to
to remove the relevant lesions while maximally pre- lower levels of the VHL protein, c-MET seems to be
serving renal parenchyma. Of course, there is a limit overexpressed in type 1 papillary tumors. The gene
to the number of procedures that can be performed on codes for a transmembrane tyrosine kinase, which
a single kidney and ‘‘completion nephrectomies’’ are acts as a receptor for hepatocyte growth factor. The
sometimes necessary. If the patient is discovered too mutations associated with HPRC are found on the
late for nephron-sparing approaches, a total nephrec- extracellular portion of the transmembrane protein
tomy may be necessary. where hepatocyte growth factor interacts with the
Recently, radiofrequency ablation and cryotherapy receptor. The mechanism by which the mutated
have been used because they are comparatively protein causes tumor formation, however, is still
P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051 1043

ing because the lesions are often isoechoic when they


are less than 3 cm in diameter and sonography should
not be used to monitor patients with HPRC [43].
Hereditary papillary renal cancer can be suspected
in a patient with two or more poorly enhancing renal
masses. Renal cystic disease is not usually a feature of
the disease but incidental cysts can occur in HPRC. If
the patient has a family history of renal cancer and
particularly if the cell type is papillary type I, the
presumptive diagnosis of HPRC can be made. This can
be confirmed with genetic testing of peripheral blood.

Fig. 4. Intraoperative ultrasound of a mixed solid and cystic


lesion within the kidney. Note that the mass is below the
surface of the kidney (arrows) and is not visible to the
surgeon. Intraoperative ultrasound assists the surgeon by
demonstrating the parenchyma deep to the surface.

unknown. Interestingly, alterations in the hepatocyte


growth factor receptor have also been found in VHL-
related tumors [42]. Moreover, there is some evidence
that sporadic papillary renal tumors found in the
general population that have c-MET alterations are
more biologically aggressive [42]
Unlike clear cell carcinomas of VHL that are
highly vascular on contrast-enhanced CT, the lesions
of HPRC tend to be hypovascular (Fig. 6). Indeed, if
density measurements are not obtained carefully,
some lesions may be mistaken for cysts [41,43].
The change in enhancement before and after intra-
venous contrast media can be as little as 10 to 15 HU.
This places a premium on obtaining scans of high
quality with the same technique (kilovolt [peak],
milliamp, slice thickness, field of view, and so forth)
both before and after contrast media administration.
The CT protocol used in these cases is the same one
Fig. 5. Patient with von Hippel-Lindau disease successfully
used for VHL patients, namely precontrast, arterial,
treated with laparoscopically assisted radiofrequency abla-
and venous phase postcontrast helical CT. Enhance- tion. Note a solid lesion (arrow) on the pretreatment study
ment on MR imaging after gadolinium chelate ad- (A) has become nonenhancing and smaller 6 months after
ministration is often very modest (15% to 20% over treatment with RF ablation (B). Cystic disease is also present
baseline) [30,43]. Renal sonography can be mislead- in the head of the pancreas.
1044 P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051

Treatment of HPRC renal tumors is similar to some patients. In general, the prognosis for HPRC is
VHL. It is generally believed that surgery can be considered excellent and many patients live normal
delayed until one of the tumors reaches 3 cm in lives with this condition. The radiologist, however,
diameter and that renal-preserving surgery should be should be alert to the possibility of HPRC when
attempted whenever feasible. If the tumor is found at multiple or bilateral low-density solid renal tumors
a diameter greater than 3 cm every attempt should be are seen.
made to perform a nephron-sparing procedure. If that
is not possible, however, a nephrectomy may be
required. Alternatively, minimally invasive radiofre-
quency ablation or cryoablation may be suitable in Hereditary leiomyoma renal cell carcinoma

Hereditary leiomyoma renal cell carcinoma


(HLRCC) is an autosomal-dominant genodermatosis
that causes cutaneous leiomyomas, uterine leiomyo-
mas, and type II papillary renal cancers. The syn-
drome, originally described in Finland among families
with hereditary uterine leiomyoma, has now been
seen throughout Europe and North America. The
association of cutaneous and uterine leiomyomas is
known as Reed’s syndrome but the association with
renal tumors is only recent [44]. The hallmarks of
HLRCC are (1) cutaneous leiomyomas over the trunk
and extremities and more rarely the face; (2) uterine
fibroids at an early age (< 30 years); and (3) type II
papillary renal tumors.
The gene for HLRCC is found on the first
chromosome (1q42.3) and is known as fumarate
hydratase. Interesting, this enzyme is a critical step
in the Krebs tricarboxylic acid cycle but its role in
causing renal tumors is not understood. Fumarate
hydratase likely acts as a tumor suppressor because
fumarate hydratase enzyme activity is low or absent
in tumors found in HLRCC [45].
Type II papillary renal cancers are found in about
17% of individuals with HLRCC and can be clini-
cally aggressive (Fig. 7). Metastases are seen in over
half of cases even with relatively small primary
tumors. HLRCC renal tumors differ from the other
hereditary renal cancer syndromes in several impor-
tant ways. Histologically, they appear to be type II
papillary tumors except for the occasional collecting
duct renal cancer. The renal tumors in HLRCC are
usually solitary and unilateral as opposed to the other
syndromes, where the tumors are usually multiple
and bilateral. The tumors are also substantially more
aggressive with Furman nuclear grades of 3 or 4 in all
cases reported and a tendency to metastasize even
when small (see Fig. 7). In contrast, the tumors
associated with VHL and HPRC are typically only
Fig. 6. (A) Precontrast CT; (B) postcontrast CT. Patient with
hereditary papillary renal cancer demonstrates a poorly Furman nuclear grade 1 or 2 and these tumors rarely
enhancing mass in the right kidney. This lesion increased in metastasize when less than 3 cm in diameter. It is
CT attenuation by only 12 HU after intravenous contrast. particularly important to differentiate HLRCC from
Minimally enhancing solid tumors are typical of the tumors HPRC. Whereas one might follow a patient with
found in HPRC. HPRC with watchful waiting, treatment is more
P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051 1045

urgent in patients with HLRCC. Tumors should be


removed when they are first seen.
Uterine fibroids occur in over 90% of women with
HLRCC. Most of these women also had skin leio-
myomas. Almost half the women with HLRCC
require a hysterectomy by the age of 30 (see Fig. 7).
In addition to screening for renal cancers patients
should be screened for uterine leiomyomas and
leiomyosarcomas. It is thought that this entity has a
higher frequency of transformation to malignant
leiomyosarcoma within the uterus, although criteria
for distinguishing these transformations by imaging
are lacking [45]. Although the cutaneous leiomyomas
do not degenerate into malignancies in general, a few
cases of cutaneous leiomyosarcomas have been
reported [45]. The cutaneous manifestations become
more prominent with age; they are hardly noticeable
when the patients are young but can become a cos-
metic issue when the patient reaches 30 to 40 years
of age.
CT is used to screen for HLRCC renal cancers and
assess the status of the uterus. Interestingly, although
the tumors tend to be hypovascular like type 1
papillary tumor, they are much more lethal. Careful
screening for metastatic disease should be performed.
MR imaging and ultrasound are suitable substitutes
when contrast-enhanced CT cannot be performed.
MR imaging of the uterus is particularly helpful in
detecting and characterizing uterine leiomyomas.

Birt-Hogg-Dubé syndrome

Birt-Hogg-Dubé syndrome (BHD), an autosomal-


dominant disorder, was originally described as a
dermatologic disorder characterized by fibrofollicu-
lomas (growths in the hair follicles) of the face and Fig. 7. Hereditary leiomyoma renal cell carcinoma. (A) A
trunk [46]. Later it became clear that there were other small mass (3 cm) is present in the middle portion of the left
markers for the disease including pulmonary cysts kidney. It is poorly enhancing typical of papillary renal
and renal tumors [47,48]. The pulmonary cysts vary cancer; however, in addition there is a metastatic lympha-
in severity and size from one or two small, scattered denopathy (arrow) adjacent to the aorta. This proved to be a
cysts to severe cystic disease complicated by sponta- papillary type II renal cancer. The renal lesion was not
visible on ultrasound. (B) Abdominal CT in another patient
neous pneumothoraces, which may be refractory to
(aged 32) with HLRCC demonstrates multiple enhancing
conventional pleurodesis. Between 15% and 30% of
leiomyomas within the uterus.
patients with BHD develop renal cancers. The renal
tumors seen in BHD are commonly, but not always,
chromophobe carcinomas or oncocytomas. Both clear
cell and papillary tumors have also been seen in BHD but the risk for colon cancer has not been clearly
[34]. Approximately 34% of the tumors are charac- defined [49,50].
terized as chromophobe carcinomas and about 50% The gene for BHD is located at 17p11.2 and codes
are hybrid chromophobe-oncocytomas. The remain- for a protein named folliculin [48,49]. Little is known
der are oncocytomas (5%); clear cell carcinoma (9%); of the mechanism of tumor formation or the function
and papillary renal cancer (2%) [34]. A higher rate of of folliculin. It is thought that this gene acts as a
colonic polyps have been observed in some families tumor suppressor and acts as a structural protein in
1046 P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051

the lung where it may form a component of the strate moderate uniform enhancement on CT, which
cytoskeletal network. This might explain the tend- is different in appearance from VHL-associated
ency of BHD patients to develop lungs cysts. The tumors, which typically have cystic components.
Hornstein-Knickenberg syndrome overlaps BHD and Birt-Hogg-Dubé syndrome is managed in a man-
is now considered to be part of BHD [51]. BHD ner similar to VHL. Tumors are generally observed
differs from other forms of hereditary renal cancer in until they reach 2 to 3 cm diameter, whereupon
that it produces a variety of cell types of renal cancer, nephron-sparing surgery is performed. Chromophobe
not just one but chromophobe cancers and their carcinomas tend to be highly enhancing and are
variants predominate. The chromophobe tumors and relatively homogeneous in appearance. Radiofre-
oncocytomas also arise from the distal renal tubules quency ablation or cryotherapy also can be consid-
in contrast to VHL and HPRC-related tumors that ered, although little outcome data are yet available.
arise from the proximal tubules [48].
The imaging of BHD should always include scans
of the lungs and abdomen (Fig. 8). The pulmonary Familial renal oncocytoma
cysts are generally found in the lower lobes and vary
in size and number from a few to multiple. Pneumo- Familial renal oncocytoma (FRO) is an incom-
thoraces may be seen despite the absence of symp- pletely characterized condition in which affected
toms. Although severe cystic disease is present in individuals develop renal oncocytomas [8]. The term
some individuals the patients do not usually become ‘‘familial’’ is used instead of ‘‘hereditary’’ to denote
oxygen-dependent. This differs from the cystic lung that a clear hereditary pattern has not yet been
disease found in lymphangioleiomyomatosis asso- established. Five families with a hereditary predispo-
ciated with tuberous sclerosis (TS, see later). The sition to renal oncocytomas were first described
chromophobe or mixed cell types typically demon- by Weirich et al in 1998 [52]. Some families had

Fig. 8. Birt-Hogg-Dube syndrome. (A) Contrast-enhanced CT demonstrates an enhancing mass in the right kidney typical of a
chromophobe carcinoma. (B) Additional smaller lesion is present in the lower pole. (C) Pulmonary CT demonstrates multiple
cysts and a loculated pneumothorax (arrow). This patient had experienced multiple pneumothoraces as a consequence of cystic
lung disease.
P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051 1047

extensive bilateral disease, and compromised renal because the sickle cell trait produces only mild
function [11]. Other families had mild manifestations symptoms and the youth of the patients (median
of FRO. There may be some overlap with BHD age about 20 years) makes it unlikely to be the result
because several families initially considered to have of a chronic process. The rate of renal tumor develop-
FRO proved to have features of BHD. Renal dys- ment even among people with sickle cell trait is low.
function without extensive neoplastic disease was Approximately 1 in 12 blacks have Hb AS (sickle cell
also noted in some family members. The prevalence trait) and relatively few reports of medullary carci-
of this entity is unknown and no putative genetic noma of the kidney have been published [56]. The
locus has yet been identified. risk of developing medullary carcinoma of the kidney
The diagnosis is based on the identification of even in the presence of sickle cell trait is negligible.
multiple oncocytomas in one or more family mem- The sickle cell gene is located at 11p15.
bers. By imaging, the lesions are indistinguishable Unfortunately, the tumors at presentation tend to
from malignant renal cancers and must be treated as if be large and are often metastatic. The tumor is
they were renal cancers (Fig. 9) [53]. When onco- generally advanced by the time it is discovered;
cytomas are extensive and confluent the term ‘‘renal median survival from the time of diagnosis is only
oncocytosis’’ can be applied [11]. Because renal 15 weeks [57]. Surgery seems to be useful only in
function is often compromised these patients are providing palliation.
often scanned with MR imaging using gadolinium- The tumors are generally large, central, and hetero-
based contrast agents. Lifelong monitoring with geneous in character. There is often evidence of ade-
imaging studies is recommended, although compli- nopathy or pulmonary metastases. These tumors
ance is lower because the perceived risk by the should be evaluated with MR imaging or CT before
patient is reduced. surgery to provide accurate clinical staging (nodal
and inferior vena caval involvement). Systemic thera-
pies are recommended but have failed significantly to
Medullary carcinoma modify the course of the illness.

Medullary carcinoma of the kidney is a rare


aggressive neoplasm that develops in young, black Other syndromes
patients (age range, 11 to 39 years) with sickle cell
trait [54]. This has led some observers to comment Tuberous sclerosis
that the renal tumors may be a secondary complica-
tion of sickle cell trait [55]. This seems doubtful Tuberous sclerosis is a genetic disease character-
ized by hamartomas in the skin, brain, and viscera.
Although not technically considered a cancer syn-
drome, TS is associated with an increased risk of
renal malignancy. TS has prevalence in the popula-
tion of approximately 1:10,000. The most common
manifestations of TS in the kidneys are cysts and
angiomyolipomas. Approximately a third of angio-
myolipomas may not contain fat visible by CT and
are difficult to differentiate from cancer. Most often
the mass represents a nonfatty angiomyolipoma.
Approximately 1% to 2% of patients with TS develop
renal cancers, which is substantially higher than the
expected rate of renal cancer in the general popula-
tion [58,59].
There is a complex relationship between renal
cancer and TS. Renal cancers are found with in-
creased frequency in patients with TS compared with
the general population and have been identified with
Fig. 9. Hereditary renal oncocytoma. Bilateral renal oncocy- mutations in both TSC1 and TSC2, the two gene loci
tomas are present in this patient. Note that the lesions are associated with TS [59,60]. Occasionally, renal can-
homogeneously enhancing. Stellate central scars, seen com- cers are even the presenting sign of TS [61]. Multi-
monly in sporadic oncocytomas, are unusual in this condition. focal renal cancer has been found in siblings from a
1048 P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051

single family with TSC1 [62]. Features indicating an Summary


association with renal cancer include a striking
female predominance (81% female versus 70% male Over the past 5 years there have been dramatic
predominance for sporadic renal cancers); median developments in the extent of knowledge of heredi-
age of only 28 years (versus sixth and seventh tary renal cancers. In addition to VHL, which is
decades for sporadic renal cancers); multifocality; associated with clear cell carcinoma, one can now
and bilaterality (43%) [63,64]. Supportive evidence list HPRC (associated with type I papillary renal
comes from the animal model of TS, the Eker rat, cancer) and HLRCC (associated with type II papillary
which has an insertional mutation in the rat TSC2 renal cancer). BHD and FRO are associated with
gene. The Eker rat develops tumors (adenomas and chromophobe carcinoma and oncocytomas, although
carcinomas) and cysts in the kidney [65]. A variety of other histologic tumor types have been found in
cell types of renal cancers have been reported in BHD. Medullary carcinoma of the kidney is asso-
humans with TS including clear cell (most common) ciated with sickle cell trait. Although the genes
papillary and chromophobe carcinomas [66,67]. associated with these tumors have been discovered,
Oncocytomas have also been reported with increased the exact mechanisms by which they cause renal
frequency in TS [68]. Some doubt has been raised cancer remain to be elucidated. It is quite likely that
concerning the actual origin of some renal tumors in other genes also are involved in this process. Using
TS because some lesions may actually be malignant VHL as an example, research is now underway on
epithelioid angiomyolipomas, which can mimic renal targeting mutant pVHL or excess HIF for diagnostic
cancers [69]. and therapeutic purposes. Understanding the mecha-
nisms leading to cancer may open new targets of
opportunity for drug development. This improved
Translocation of chromosome 3 knowledge of the biogenetic pathways used to form
tumors will impact the development of new thera-
A number of families have been reported in which peutic techniques for treating renal cancers in heredi-
part of the short arm of chromosome 3 has been tary and nonhereditary forms of the disease.
translocated to another chromosome [9,17]. These
balanced translocations predispose the patient to the
development of clear cell carcinomas of the kidney. References
Because VHL is also located on chromosome 3 it was
originally assumed that this was a subset of VHL; [1] Moyad MA. Review of potential risk factors for kidney
however, patients with chromosome 3 translocation (renal cell) cancer. Semin Urol Oncol 2001;19:280 – 93.
do not have other stigmata of VHL and the VHL [2] Gago-Dominguez M, Yuan JM, Castelao JE, Ross RK,
portion of the short arm of chromosome 3 is often Yu MC. Family history and risk of renal cell carci-
intact [70,71]. Translocation of the chromosome 3 noma. Cancer Epidemiol Biomarkers Prev 2001;10:
can result in a clear-cell predominant form of heredi- 1001 – 4.
tary renal cancer. Unlike the other syndromes, which [3] Amin MB, Tamboli P, Javidan J, Stricker H, de-Peralta
require a careful analysis of the genes for mutations, Venturina M, Deshpande A, et al. Prognostic impact
of histologic subtyping of adult renal epithelial neo-
this abnormality can be detected on a karyotype of
plasms: an experience of 405 cases. Am J Surg Pathol
peripheral white blood cells. 2002;26:281 – 91.
[4] Bostwick DG, Eble JN. Diagnosis and classification of
renal cell carcinoma. Urol Clin North Am 1999;26:
Familial renal cancer 627 – 35.
[5] Storkel S, Eble JN, Adlakha K, Amin M, Blute ML,
Not all families with tendencies to develop renal Bostwick DG, et al. Classification of renal cell carci-
cancers are explained by the syndromes described noma: Workgroup No. 1. Union Internationale Contre
previously. As a result, the term ‘‘familial renal le Cancer (UICC) and the American Joint Committee
cancer’’ is used to characterize patients with apparent on Cancer (AJCC). Cancer 1997;80:987 – 9.
[6] Kovacs G, Akhtar M, Beckwith BJ, Bugert P, Cooper
genetic predisposition to renal cancer in whom the
CS, Delahunt B, et al. The Heidelberg classification
exact gene involved has not yet been determined. of renal cell tumours. J Pathol 1997;183:131 – 3.
Fortunately, as more is learned about the origins of [7] Nagashima Y. Chromophobe renal cell carcinoma:
hereditary renal cancer this category contains fewer clinical, pathological and molecular biological aspects.
patients and more are successfully characterized by Pathol Int 2000;50:872 – 8.
specific diagnoses. [8] Erlandson RA, Shek TW, Reuter VE. Diagnostic sig-
P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051 1049

nificance of mitochondria in four types of renal epithe- the renal pathology of a familial form of renal cell
lial neoplasms: an ultrastructural study of 60 tumors. carcinoma associated with von Hippel-Lindau disease:
Ultrastruct Pathol 1997;21:409 – 17. clinical and molecular genetic implications. J Urol
[9] Amin MB, Crotty TB, Tickoo SK, Farrow GM. Renal 1995;153: 22 – 6.
oncocytoma: a reappraisal of morphologic features [24] Choyke PL, Glenn GM, Walther MM, Patronas NJ,
with clinicopathologic findings in 80 cases. [published Linehan WM, Zbar B. von Hippel-Lindau disease:
erratum appears in Am J Surg Pathol 1997;21:742] genetic, clinical, and imaging features. Radiology
[see comments] Am J Surg Pathol 1997;21:1 – 12. 1995;194:629 – 42.
[10] Noguchi S, Nagashima Y, Shuin T, Kubota Y, Kita- [25] Choyke PL, Glenn GM, Walther MM, Zbar B, Weiss
mura H, Yao M, et al. Renal oncocytoma containing GH, Alexander RB, et al. The natural history of renal
‘‘chromophobe’’ cells. Int J Urol 1995;2:279 – 80. lesions in von Hippel-Lindau disease: a serial CT study
[11] Warfel KA, Eble JN. Renal oncocytomatosis. J Urol in 28 patients. AJR Am J Roentgenol 1992;159:
1982;127:1179 – 80. 1229 – 34.
[12] Figenshau RS, Basler JW, Ritter JH, Siegel CL, Simon [26] Turner KJ, Huson SM, Moore N, Britton BJ, Cranston
JA, Dierks SM. Renal medullary carcinoma. J Urol D. Von Hippel-Lindau disease: renal tumors less than
1998;159:711 – 3. 3 cm can metastasize. J Urol 2001;165:1207.
[13] Davidson AJ, Choyke PL, Hartman DS, Davis Jr. CJ. [27] Herring JC, Enquist EG, Chernoff A, Linehan WM,
Renal medullary carcinoma associated with sickle cell Choyke PL, Walther MM. Parenchymal sparing sur-
trait: radiologic findings. Radiology 1995;195:83 – 5. gery in patients with hereditary renal cell carcinoma:
[14] Hamano K, Esumi M, Igarashi H, Chino K, Mochida J, 10-year experience. J Urol 2001;165:777 – 81.
Ishida K, et al. Biallelic inactivation of the von Hippel- [28] Walther MM, Choyke PL, Glenn G, Lyne JC, Rayford
Lindau tumor suppressor gene in sporadic renal cell W, Venzon D, et al. Renal cancer in families with
carcinoma. J Urol 2002;167:713 – 7. hereditary renal cancer: prospective analysis of a tumor
[15] Herman JG, Latif F, Weng Y, Lerman MI, Zbar B, Liu size threshold for renal parenchymal sparing surgery.
S, et al. Silencing of the VHL tumor-suppressor gene J Urol 1999;161:1475 – 9.
by DNA methylation in renal carcinoma. Proc Natl [29] Glenn GM, Gnarra JR, Choyke PL, Walther MM, Zbar
Acad Sci U S A 1994;91:9700 – 4. B, Linehan WM. The molecular genetics of renal
[16] Latif F, Tory K, Gnarra J, Yao M, Duh FM, Orcutt ML, cell carcinoma. In: Raghaven D, Scher HI, Lange
et al. Identification of the von Hippel-Lindau disease PH, editors. Principles and practice of genitouri-
tumor suppressor gene. [see comments] Science 1993; nary oncology. Philadelphia: Lippincott-Raven; 1997.
260:1317 – 20. p. 85 – 97.
[17] Kaelin WG, Iliopoulos O, Lonergan KM, Ohh M. [30] Ho VB, Allen S, Hood M, Choyke PL. Renal masses:
Functions of the von Hippel-Lindau tumour suppressor quantitative assessment of enhancement with dynamic
protein. J Intern Med 1998;243:535 – 9. MR imaging. Radiology 2002;224:695 – 700.
[18] Bindra RS, Vasselli JR, Stearman R, Linehan WM, [31] Jamis-Dow CA, Choyke PL, Jennings SB, Linehan
Klausner RD. VHL-mediated hypoxia regulation of WM, Thakore KN, Walther MM. Small (< or =
cyclin D1 in renal carcinoma cells. Cancer Res 2002; 3-cm) renal masses: detection with CT versus US and
62:3014 – 9. pathologic correlation. Radiology 2002;224:695 – 700.
[19] Igarashi H, Esumi M, Ishida H, Okada K. Vascular [32] Choyke PL, Pavlovich CP, Daryanani KD, Hewitt SM,
endothelial growth factor overexpression is correlated Linehan WM, Walther MM. Intraoperative ultrasound
with von Hippel-Lindau tumor suppressor gene in- during renal parenchymal sparing surgery for heredi-
activation in patients with sporadic renal cell carci- tary renal cancers: a 10-year experience. J Urol 2001;
noma. Cancer 2002;95:47 – 53. 165:397 – 400.
[20] Wiesener MS, Seyfarth M, Warnecke C, Jurgensen JS, [33] Wood BJ, Ramkaransingh JR, Fojo T, Walther MM,
Rosenberger C, Morgan NV, et al. Paraneoplastic Libutti SK. Percutaneous tumor ablation with radiofre-
erythrocytosis associated with an inactivating point quency. Cancer 2002;94:443 – 51.
mutation of the von Hippel-Lindau gene in a renal cell [34] Pavlovich CP, Walther MM, Eyler RA, Hewitt SM,
carcinoma. Blood 2002;99:3562 – 5. Zbar B, Linehan WM, et al. Renal tumors in the
[21] Richard S, Croisille L, Yvart J, Casadeval N, Eschwege Birt-Hogg-Dube syndrome. Am J Surg Pathol 2002;
P, Aghakhani N, et al. Paradoxical secondary poly- 26:1542 – 52.
cythemia in von Hippel-Lindau patients treated [35] Gervais DA, McGovern FJ, Wood BJ, Goldberg SN,
with anti-vascular endothelial growth factor receptor McDougal WS, Mueller PR. Radio-frequency ablation
therapy. Blood 2002;99:3851 – 3. of renal cell carcinoma: early clinical experience.
[22] Stolle C, Glenn G, Zbar B, Humphrey JS, Choyke P, Radiology 2000;217:665 – 72.
Walther M, et al. Improved detection of germline [36] Chen RN, Novick AC, Gill IS. Laparoscopic cryoabla-
mutations in the von Hippel-Lindau disease tumor tion of renal masses. Urol Clin North Am 2000;27:
suppressor gene. Hum Mutat 1998;12:417 – 23. 813 – 20.
[23] Poston CD, Jaffe GS, Lubensky IA, Solomon D, Zbar [37] Pautler SE, Pavlovich CP, Mikityansky I, Drachenberg
B, Solomon D, Linehan WM, et al. Characterization of DE, Choyke PL, Linehan WM, et al. Retroperito-
1050 P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051

neoscopic-guided radiofrequency ablation of renal tu- [51] Schulz T, Hartschuh W. Birt-Hogg-Dube syndrome
mors. Can J Urol 2001;8:1330 – 3. and Hornstein-Knickenberg syndrome are the same:
[38] Remer EM, Weinberg EJ, Oto A, O’Malley CM, Gill different sectioning technique as the cause of different
IS. MR imaging of the kidneys after laparoscopic cryo- histology. J Cutan Pathol 1999;26:55 – 61.
ablation. AJR Am J Roentgenol 2000;174:635 – 40. [52] Weirich G, Glenn G, Junker K, Merino M, Storkel S,
[39] Amin MB, Corless CL, Renshaw AA, Tickoo SK, Lubensky I, et al. Familial renal oncocytoma: clinico-
Kubus J, Schultz DS. Papillary (chromophil) renal cell pathological study of 5 families. J Urol 1998;160:
carcinoma: histomorphologic characteristics and eval- 335 – 40.
uation of conventional pathologic prognostic parame- [53] Davidson AJ, Hayes WS, Hartman DS, McCarthy
ters in 62 cases. [see comments] Am J Surg Pathol CJ, Davis Jr. CJ. Renal oncocytoma and carcinoma:
1997;21:621 – 35. failure of differentiation with CT. Radiology 1993;186:
[40] Inoue K, Karashima T, Chikazawa M, Iiyama T, Yosh- 693 – 6.
ikawa C, Furihata M, et al. Overexpression of c-met [54] Davis Jr. CJ, Mostofi FK, Sesterhenn IA. Renal medul-
proto-oncogene associated with chromophilic renal cell lary carcinoma: the seventh sickle cell nephropathy.
carcinoma with papillary growth. Virchows Arch 1998; Am J Surg Pathol 1995;19:1 – 11.
433:511 – 5. [55] Ataga KI, Orringer EP. Renal abnormalities in sickle
[41] Schmidt L, Junker K, Weirich G, Glenn G, Choyke P, cell disease. Am J Hematol 2000;63:205 – 11.
Lubensky I, et al. Two North American families with [56] Ashley-Koch A, Yang Q, Olney RS. Sickle hemoglo-
hereditary papillary renal carcinoma and identical bin (HbS) allele and sickle cell disease: a HuGE
novel mutations in the MET proto-oncogene. Cancer review. Am J Epidemiol 2000;151:839 – 45.
Res 1998;58:1719 – 22. [57] Pickhardt PJ. Renal medullary carcinoma: an aggres-
[42] Koochekpour S, Jeffers M, Wang PH, Gong C, Taylor sive neoplasm in patients with sickle cell trait. Abdom
GA, Roessler LM, et al. The von Hippel-Lindau tumor Imaging 1998;23:531 – 2.
suppressor gene inhibits hepatocyte growth factor/scat- [58] Chan HS, Daneman A, Gribbin M, Martin DJ. Renal
ter factor-induced invasion and branching morphogen- cell carcinoma in the first two decades of life. Pediatr
esis in renal carcinoma cells. Mol Cell Biol 1999;19: Radiol 1983;13:324 – 8.
5902 – 12. [59] Bjornsson J, Short MP, Kwiatkowski DJ, Henske EP.
[43] Choyke PL, Walther MM, Glenn GM, Wagner JR, Tuberous sclerosis-associated renal cell carcinoma:
Venzon DJ, Lubensky IA, et al. Imaging features of clinical, pathological, and genetic features. Am J Pathol
hereditary papillary renal cancers. J Comput Assist 1996;149:1201 – 8.
Tomogr 1997;21:737 – 41. [60] Robertson FM, Cendron M, Klauber GT, Harris BH.
[44] Launonen V, Vierimaa O, Kiuru M, Isola J, Roth S, Renal cell carcinoma in association with tuberous scle-
Pukkala E, et al. Inherited susceptibility to uterine rosis in children. J Pediatr Surg 1996;31:729 – 30.
leiomyomas and renal cell cancer. Proc Natl Acad [61] Kulkarni S, Uddar M, Deshpande SG, Vaid S, Wadia
Sci U S A 2001;98:3387 – 92. RS. Renal cell carcinoma as significant manifestation
[45] Tomlinson IP, Alam NA, Rowan AJ, Barclay E, Jaeger of tuberous sclerosis complex. J Assoc Physicians
EE, Kelsell D, et al. Germline mutations in FH predis- India 2000;48:351 – 3.
pose to dominantly inherited uterine fibroids, skin leio- [62] Sampson JR, Patel A, Mee AD. Multifocal renal cell
myomata and papillary renal cell cancer. Nat Genet carcinoma in sibs from a chromosome 9 linked (TSC1)
2002;30:406 – 10. tuberous sclerosis family. J Med Genet 1995;32:
[46] Birt AR, Hogg GR, Dube WJ. Hereditary multiple 848 – 50.
fibrofolliculomas with trichodiscomas and acrochor- [63] Torres VE, Zincke H, King BK, Bjornsson J. Renal
dons. Arch Dermatol 1977;113:1674 – 7. manifestations of tuberous sclerosis complex. Contrib
[47] Roth JS, Rabinowitz AD, Benson M, Grossman ME. Nephrol 1997;122:64 – 75.
Bilateral renal cell carcinoma in the Birt-Hogg-Dube [64] Washecka R, Hanna M. Malignant renal tumors in
syndrome. J Am Acad Dermatol 1993;29:1055 – 6. tuberous sclerosis. Urology 1991;37:340 – 3.
[48] Nickerson ML, Warren MB, Toro JR, Matrosova V, [65] Eker R, Mossige J, Johannessen JV, Aars H. Hereditary
Glenn G, Turner ML, et al. Mutations in a novel gene renal adenomas and adenocarcinomas in rats. Diagn
lead to kidney tumors, lung wall defects and benign Histopathol 1981;4:99 – 110.
tumors of the hair follicle in patients with Birt-Hogg- [66] Hidai H, Chiba T, Takagi Y, Taki A, Nagashima Y,
Dube syndrome. Cancer Cell 2002;2:157 – 64. Kuroko K. Bilateral chromophobe cell renal carcinoma
[49] Khoo SK, Giraud S, Kahnoski K, Chen J, Motorna O, in tuberous sclerosis complex. Int J Urol 1997;4:86 – 9.
Nickolov R, et al. Clinical and genetic studies of Birt- [67] Peccatori I, Pitingolo F, Battini G, Meroni M, Gior-
Hogg-Dube syndrome. J Med Genet 2002;39:906 – 12. dano F, Guarino M, et al. Pulmonary lymphangioleio-
[50] Zbar B, Alvord WG, Glenn G, Turner M, Pavlovich myomatosis and renal papillary cancer: incomplete
CP, Schmidt L, et al. Risk of renal and colonic neo- expression of tuberous sclerosis? Nephrol Dial Trans-
plasms and spontaneous pneumothorax in the Birt- plant 1997;12:2740 – 3.
Hogg-Dube syndrome. Cancer Epidemiol Biomarkers [68] Jimenez RE, Eble JN, Reuter VE, Epstein JI, Folpe AL,
Prev 2002;11:393 – 400. de Peralta-Venturina M, et al. Concurrent angiomyoli-
P.L. Choyke / Radiol Clin N Am 41 (2003) 1037–1051 1051

poma and renal cell neoplasia: a study of 36 cases. Mod SC, et al. Hereditary renal-cell carcinoma associated
Pathol 2001;14:157 – 63. with a chromosomal translocation. N Engl J Med
[69] Pea M, Bonetti F, Martignoni G, Henske EP, Manfrin 1979;301:592 – 5.
E, Colato C, et al. Apparent renal cell carcinomas in [71] Bodmer D, Eleveld MJ, Ligtenberg MJ, Weterman MA,
tuberous sclerosis are heterogeneous: the identification Janssen BA, Smeets DF, et al. An alternative route for
of malignant epithelioid angiomyolipoma. Am J Surg multistep tumorigenesis in a novel case of hereditary
Pathol 1998;22:180 – 7. renal cell cancer and a t(2;3)(q35;q21) chromosome
[70] Cohen AJ, Li FP, Berg S, Marchetto DJ, Tsai S, Jacobs translocation. Am J Hum Genet 1998;62:1475 – 83.
Radiol Clin N Am 41 (2003) 1053 – 1065

Surgical management of renal tumors


Rizk El-Galley, MB Bch, FRCS
Department of Surgery, Division of Urology, University of Alabama at Birmingham, 1530 3rd Avenue South, MEB 602,
Birmingham, AL 35294 – 3296, USA

Renal cell carcinoma is a relatively rare tumor, than 5% of all renal cell carcinomas have a cystic
accounting for approximately 3% of malignancies in appearance with septations, irregular borders, dystro-
adults, but is the most common tumor of the kidney phic calcification, or other features that distinguish it
and the third most common tumor seen by urologists from a simple renal cyst.
[1]. The classic symptom triad of pain, hematuria, The differential diagnosis of solid kidney masses
and flank pain is certainly a reliable clinical symptom includes oncocytoma (granular oncocytes on histo-
complex. More recently, however, most renal cell logic analysis, with a central scar in the tumor);
carcinomas are diagnosed at earlier stages and are angiomyolipoma (contains fat, seen on CT scans);
frequently found incidentally at radiologic investiga- xanthogranulomatous pyelonephritis (usually in pa-
tion done for other reasons [2]. tients with diabetes, with a concurrent stone in a
Renal cell carcinoma is refractory to most tradi- poorly functioning kidney); fibromas; or metastasis.
tional oncologic treatments, including chemotherapy, Despite the diagnostic clues seen at radiologic inves-
radiation therapy, and hormonal therapy [3 – 5]. Radi- tigation, the histologic nature of these masses cannot
cal nephrectomy, removing all the contents of Ge- be confirmed without tissue biopsy, which is gener-
rota’s fascia, is considered the standard treatment for ally avoided because of the risk for seeding malignant
localized tumors. More recent data indicate, however, cells through the needle track or the possibility of
that in carefully selected patients partial nephrectomy obtaining benign tissue approximating a malignant
may be an option [6 – 8]. The role of radical nephrec- area. Accordingly, in most of these patients radical
tomy in patients with metastatic disease is contro- nephrectomy is required before the kidney lesion is
versial and is not indicated unless the patient has finally diagnosed pathologically.
intractable bleeding or pain, or it is necessary to debulk CT and MR imaging are the imaging studies most
the tumor for immunotherapy or other systemic thera- commonly used to stage renal tumors. Abdominal CT
pies. Local extension into the renal vein or inferior is particularly useful to show local extension of tumor
vena cava (IVC) is not considered a contraindication to and the presence of enlarged para-aortic lymph nodes.
radical nephrectomy. Tumor extension beyond Gero- MR imaging is superior to CT for determining the
ta’s fascia involving other organs is associated with superior extent of a vena caval thrombus; however,
poor prognosis, however, and nephrectomy should be the new generation of CT scanners with rapid image
considered only for palliation or as part of an adjuvant acquisition are as accurate as MR imaging in vena
therapy protocol [9 – 12]. caval imaging [13,14]. These new imaging studies
Because of recent advances in sophisticated ra- have replaced, to large extent, venocavography and
diologic studies, the surgeon can now make an arteriography, which are more invasive. Chest radiog-
accurate preoperative assessment of the nature and raphy or chest CT is routinely done to rule out
extent of kidney tumors. The diagnosis of renal cell pulmonary metastasis; bone scanning is required only
carcinoma is generally made with CT, showing a in the presence of a large tumor or if clinical evalua-
solid mass in the parenchyma of the kidney. Fewer tion suggests metastasis to bone.
When evaluating renal tumors, the urologist is
E-mail address: rizk.el-galley@ccc.uab.edu looking for certain information to help in constructing

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00077-0
1054 R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065

a management plan. Here are some of the points that Surgical planning is usually dependent on radio-
contribute in the surgical decision-making. logic information. The extent of the tumor and its
location in the kidney, proximity to the renal collecting
system and renal vessels, and presence of fat planes
Is it a kidney a tumor or a pseudotumor? between the tumor and other structures (eg, liver,
colon, and posterior abdominal wall muscles) are all
Pseudotumors in the kidney are rare; however, this important information that help the surgeon to assess
diagnosis should be taken into consideration. A the local invasiveness of the tumor. Similarly, tumor
hypertrophied column of Bertin, inflammatory renal extension into the renal vein or IVC and the level of
mass, or perinephric inflammation extending to the this extension are essential information in surgical
kidney may be confused with a renal tumor. A CT decision-making. Tumors that extend beyond the distal
scan is usually very helpful in delineating the nature two thirds of the renal vein are not suitable for
of these tumors. In some cases, when the nature of the laparoscopic surgery. The level of extension in the
mass in undetermined, a repeat CT a few weeks later IVC should be assessed accurately before surgery.
shows a significant change in an inflammatory mass, Tumors that extend above the hepatic veins require
whereas a tumor change is less remarkable. A hyper- full mobilization of the liver to control the IVC above
trophied column of Bertin has been a diagnostic the hepatic veins. It also requires the Pringle maneuver
problem on intravenous pyelogram and a dimercap- (clamping the porta hepatis), clamping the renal veins
tosuccinic acid (DMSA) nuclear scan was required to and the lumbar veins to reduce blood loss during tumor
establish the diagnosis; a column of Bertin is homog- extraction from the IVC. Tumors that extend into the
enous with the rest of the kidney tissue, whereas a right atrium require the involvement of a cardiotho-
tumor shows different isotope uptake. Most of these racic surgeon and cardiopulmonary bypass to extract
swellings, however, can be differentiated with a CT the tumor from the IVC and atrium. The presence of
scan that obviates the need for a nuclear scan [15,16]. intra-abdominal metastasis, the function of the contra-
lateral kidney, and the appearance of the adrenal glands
are valuable information for surgical decision making.
Is it a cystic tumor?

Cystic renal masses range from a simple cyst to Surgical anatomy of the kidneys
cystic renal carcinoma. Characterization of cystic
renal masses relies mainly on the Bosniak classifica- The kidneys are paired, reddish brown, solid
tion, which consists of four categories [17 – 19]: organs situated on each side of the midline in the
benign simple cysts (category I); minimally compli- retroperitoneal space. Their weight depends on body
cated cysts (category II); indeterminate cystic renal size, averaging 150 and 135 g each in the adult man
masses that include cystic renal tumors (multilocu- and woman, respectively. Kidneys in mature adults
lated or not) and complex cysts (category III); and vary in length from 11 to 14 cm, in width from 5 to
cystic renal cell carcinomas (category IV). Usually, 7 cm, and in thickness from 2.5 to 3 cm. Because of
category I cysts are not indication for surgery unless the effect of the hepatic mass, the right kidney is
they are symptomatic, category II are most likely shorter and broader and lays 1 to 2 cm lower than the
benign and can be watched, category III are more left kidney.
likely to be malignant, and category IV are highly Each kidney is surrounded by a layer of fat, covered
suspicious for being malignant [20,21]. by the Gerota’s fascia (Fig. 1). Gerota’s fascia is
completely fused above and lateral to the kidney;
medially and inferiorly fusion is incomplete. This
Is it a solid renal tumor? incomplete fusion is of clinical importance in deter-
mining the possible routes of spread of bleeding or
In general, solid renal tumors should be consid- infection around the kidneys. Both layers of Gerota’s
ered malignant until proved otherwise, with the fascia probably continue across the midline, with the
exception of a tumor that contains fat on the CT posterior layer crossing behind the great vessels and
scanning (angiomyolipoma) or tumors that do not the anterior layer extending in front of the great
enhance on a CT and do not grow on follow-up CTs. vessels. The parietal peritoneum fuses with the anterior
Oncocytoma is another tumor that is difficult to layer of Gerota’s fascia to form the white line of Toldt
differentiate from malignant tumors radiologically laterally. During surgical approaches to the kidneys,
and is usually diagnosed after excision [22]. incision along this line enables the surgeon to reflect
R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065 1055

Fig. 1. Gerota’s fascia. (From El-Galley RES, Keane TE. Kidneys, ureters, and bladders. In: Wood WC, Skandalakis JE, editors.
Anatomic basis of tumor surgery. St. Louis: Quality Medical Publishing; 1999; with permission.)

the peritoneum with the mesocolon through a relative- kidney lies on the diaphragm, behind which is the
ly bloodless plane and gives access to the renal hilum. pleural reflection. An operative approach to this area
The upper pole of the left kidney lies at the level with a high incision above the eleventh or tenth rib
of the twelfth thoracic vertebral body and the lower risks entering the pleural space. The upper border of
pole at the level of the third lumbar vertebra (Fig. 2). the left kidney usually extends to the upper border of
The right kidney usually extends from the top of the the eleventh rib, and the upper pole of the right
first lumbar vertebra to the bottom of the third lumbar kidney, which is lower, is usually at the level of the
vertebra. Because of the free mobility of the kidneys, eleventh intercostal space. The lower two thirds of the
these relationships change with both body position posterior surface of both kidneys lies on three
and respiration. muscles, which from medial to lateral are the psoas
The right adrenal gland covers the uppermost part major, quadratus lumborum, and the aponeurosis of
of the anteromedial surface of the right kidney. The the transversus abdominis muscles. The renal vessels
anterior relationships of the right kidney include and pelvis lie against the contour of the psoas muscle,
the liver, which overlies the upper two thirds of the which tilts the lower pole of each kidney away from
anterior surface, and the hepatic flexure of the colon, the midline. Alterations in this alignment may be seen
which overlies the lower third. The right renal hilum with space-occupying lesions and should prompt
is overlaid by the second part of the duodenum. The careful assessment.
anterior surface of the kidney beneath the liver is the The renal parenchyma is divided into an internal
only area covered by peritoneum. The anteromedial darker medulla and an external lighter-hued cortex
surface of the left kidney is also covered by the left (Fig. 3). The medulla is composed of 8 to 18 conical
adrenal gland in its uppermost part. The spleen, body structures called the ‘‘renal pyramids,’’ which are
of the pancreas, stomach, and splenic flexure of the made of ascending and descending loops of Henle
colon are all anterior to the left kidney. The area of and collecting ducts. The round tip of each pyramid is
the kidney beneath the small intestine, the spleen, and known as the ‘‘renal papilla.’’ These papillae cannot be
the stomach is covered by peritoneum. Both kidneys seen during surgical dissections because each papillary
share relatively symmetric relations to the posterior projection is encompassed by a smooth muscular
abdominal wall. The upper third or upper pole of each sleeve called a ‘‘minor calyx.’’ These minor calyces
1056 R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065

Fig. 2. Location of kidneys. (From El-Galley RES, Keane TE. Kidneys, ureters, and bladders. In: Wood WC, Skandalakis JE,
editors. Anatomic basis of tumor surgery. St. Louis: Quality Medical Publishing; 1999; with permission.)

coalesce to form two or three major calyces, which in Blood supply


turn join to form the renal pelvis. The renal pelvis
extends through the renal hilum behind the renal Each kidney is classically supplied by a renal
vessels and continues as the ureter. Anatomic varia- artery and a larger renal vein, arising from the aorta
tions in the renal pelvis are not uncommon. The renal and the IVC, respectively, at the level of the second
pelvis, which is usually partially extrarenal, may lie lumbar vertebra below the takeoff of the superior
completely outside or within the kidney. Occasionally mesenteric artery (Fig. 4). These vessels enter the
the renal pelvis may be duplicated, with duplication of renal hilum medially, with the vein anterior to the
the renal units. Anatomic variations of the renal pelvis artery and both anterior to the renal pelvis. Although
tend to occur bilaterally, which should be considered the right kidney is lower than the left kidney, the right
when evaluating urographic studies to differentiate renal artery arises from the aorta at a higher level and
pathologic conditions from normal variations. takes a longer course than the left renal artery. It
The renal cortex lies between the bases of the travels downward behind the IVC to reach the right
pyramids and the renal capsule. The tongues of cortical kidney, whereas the left renal artery passes slightly
tissue that extend between the renal pyramids are upward to reach the left kidney. Because of the
called the ‘‘columns of Bertin’’ and, when enlarged, posterior position of the kidneys, both renal arteries
can closely resemble a renal mass. The outer border of course slightly posterior. Two small but important
the renal cortex should be smooth. Indentations on the branches arise from the main renal artery before its
cortical surface might represent persistent fetal lobu- termination in the hilum: the inferior adrenal artery and
lations, previous scarring, and infection or space- the artery that supplies the renal pelvis and upper
occupying lesion. ureter. Ligation of this branch may result in ischemia
R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065 1057

to the area of the upper ureter with stricture formation.


The main renal artery divides into five segmental
arteries at the renal hilum. Each segmental artery is
an end artery; occlusion leads to ischemia and infarc-
tion of the corresponding renal segment. The first
branch is the posterior artery, which arises just before
the renal hilum and passes posterior to the renal pelvis
to supply a large posterior segment of the kidney. The
main renal artery then terminates into four anterior
segmental arteries at the renal hilum: (1) the apical,
(2) upper, (3) middle, and (4) lower anterior segmental
arteries (Fig. 5). Both the apical and inferior arteries
supply the anterior and posterior surfaces of the upper
and lower poles of the kidneys, respectively. The upper
and middle arteries supply two corresponding seg-
ments on the anterior surface of the kidney. Renal
vascular segments are also identified.
The segmental arteries course though the renal
sinus and branch into the lobar arteries, which are
Fig. 3. Anatomy of renal parenchyma. (From El-Galley
usually distributed one for each pyramid. Each lobar
RES, Keane TE. Kidneys, ureters, and bladders. In: Wood
WC, Skandalakis JE, editors. Anatomic basis of tumor
artery divides into two or three interlobar arteries that
surgery. St. Louis: Quality Medical Publishing; 1999; pass between the renal pyramids to the corticomedul-
with permission.) lary junction, where they become the arcuate artery.
The arcuate arteries, as their name implies, arch over
the bases of the pyramids and give rise to a series of
interlobular arteries, which in turn take a straight
course to the renal cortex, with some terminal small
branches anastomosing with the capsular arteries. This

Fig. 4. Blood supply to the kidney (anterior surface of right kidney). (From El-Galley RES, Keane TE. Kidneys, ureters, and
bladders. In: Wood WC, Skandalakis JE, editors. Anatomic basis of tumor surgery. St. Louis: Quality Medical Publishing; 1999;
with permission.)
1058 R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065

Fig. 5. Vascular segments (left kidney). (From El-Galley RES, Keane TE. Kidneys, ureters, and bladders. In: Wood WC,
Skandalakis JE, editors. Anatomic basis of tumor surgery. St. Louis: Quality Medical Publishing; 1999; with permission.)

anastomosis can enlarge to supply a significant amount vein. These nodes form the first station for lymphatic
of blood to the superficial cortical glomeruli, particu- spread of renal cancer. On the left side, lymphatic
larly in cases of gradual narrowing of the renal arteries. trunks from the renal hilum drain to the para-aortic
The renal cortex is drained by the interlobular veins, lymph nodes from the level of the inferior mesenteric
which, unlike the renal arteries, anastomose freely with artery to the diaphragm. Lymphatic vessels from the
the arcuate veins at the base of the medullary pyramids right kidney drain into the lateral paracaval and
and with the capsular and perirenal veins on the surface interaortocaval nodes from the level of the common
of the kidney. The arcuate veins drain through the iliac vessels to the diaphragm. Lymphatic vessels
interlobar veins to the lobar veins, which join to form from both sides may extend above the diaphragm to
the renal vein. The right renal vein, 2 to 4 cm long, the retrocrural nodes or directly into the thoracic duct.
joins the lateral aspect of the IVC, usually without
receiving any tributaries. The left renal vein, 6 to 10 cm
long, crosses anterior to the aorta and ends in the left Surgical applications
aspect of the IVC. It receives three tributaries lateral to
the aorta: (1) the left adrenal vein superiorly, (2) left The kidneys can be approached through various
gonadal vein inferiorly, and (3) a lumbar vein poste- incisions: lumbar, anterior transperitoneal, thoraco-
riorly. At the renal hilum the renal vein usually lies in abdominal, and posterior lumbar. Factors that should
front of the renal artery. Passing more medially, be taken into consideration before selecting an inci-
however, the renal artery may be a centimeter higher sion include type of operation and pathologic condi-
or lower than the vein. tion, body habitus, and pulmonary or spinal
deformities. Small uncomplicated tumors can be
Lymphatic drainage approached through an extraperitoneal flank incision.
This approach has the advantages of being extraperi-
Lymphatic vessels within the renal parenchyma toneal, with a shorter period of ileus, and in obese
consist of cortical and medullary plexuses that follow patients most of the panniculus falls away from the
the renal vessels to the renal sinus and form several kidney. Exposure of the renal pedicle with lateral
large lymphatic trunks. The renal sinus is the site of lumbar approaches is not as good as an anterior
numerous communications between lymphatic ves- approach, however, and runs the risk of entering the
sels from the perirenal tissues, renal pelvis, and upper pleural cavity, particularly if a supracostal incision is
ureter. Initial, lymphatic drainage runs to the nodes performed. This incision can be performed above the
present at the renal hilum lying close to the renal twelfth or eleventh rib, either extrapleural or intra-
R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065 1059

pleural, to expose the suprarenal gland or the upper high caval thrombi. Unilateral anterior extraperitoneal
pole of the kidney, and can also be extended down- incision provides adequate exposure in noncompli-
ward to expose the ureter. cated cases. Bilateral tumors can be approached with
For good exposure of the renal vessels, particu- a midline or Chevron incision; however, such lesions
larly for operations for advanced tumors, an ante- are best approached one side at a time.
rior transperitoneal approach is preferred. It can be On the right side, once the peritoneum is entered,
performed through an anterior subcostal, midline, the intra-abdominal contents, mainly the liver, are
or paramedian incision. The midline incision is inspected for unrecognized metastasis, and the tumor
faster to perform and to close, but the incidence is examined carefully for resectability. The dia-
of incisional hernia is higher than with paramedian phragm is retracted superiorly with a self-retaining
incisions. Posterior lumbar incisions are easy to retractor, and countertraction is applied to the supe-
perform and are easier on the patient, but the rior border of the rib below after releasing the
exposure is limited, particularly with respect to costochondral ligament. The liver is kept out of the
renal vessels. Good access is provided to the renal way by gentle retraction to prevent hepatic injury.
pelvis and upper third of the ureter for stone During extensive IVC mobilization care must be
surgery, but this approach is not recommended taken not to injure the short caudate veins.
for malignancies. Attention should be given to the renal pedicle,
which can be approached ventrally by retracting the
Radical nephrectomy ascending colon and dividing the lateral paracolic
peritoneum (Fig. 6). The hepatic flexure and duode-
The eleventh or twelfth rib supracostal incision, num are mobilized medially to expose the renal
with attempt to remain extrapleural, is recommended pedicle and the renal veins lying in front of the artery.
for most cases. It provides good exposure of the As an alternative, with the dorsal approach to the
kidney, renal pedicle, and adjacent organs. Thora- renal pedicle the renal artery is readily accessible for
coabdominal incision is preferable in patients with ligation and division. This maneuver significantly
large upper pole tumors or tumors that extend into the reduces potential blood loss. It can be performed by
IVC, although median sternotomy is an option for dissecting the kidney and surrounding tissues free

Fig. 6. Approach to the renal pedicle. (From El-Galley RES, Keane TE. Kidneys, ureters, and bladders. In: Wood WC,
Skandalakis JE, editors. Anatomic basis of tumor surgery. St. Louis: Quality Medical Publishing; 1999; with permission.)
1060 R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065

from the posterior abdominal wall and rotating it is incised (Fig. 7). The mesentery is dissected bluntly
medially, after which the renal artery can be identi- from the anterior surface of Gerota’s fascia. Care
fied, ligated, and divided. should be taken to prevent injury to the pancreas,
The ureter, gonadal vessels, and periureteral fat which is mobilized medially. If the tumor extends
are dissected free of the posterior peritoneum and into the colonic mesentery, this part of the mesentery
divided in two or three separate bundles. The dissec- can be resected with the specimen without great risk
tion is then carried superiorly along the IVC on its for colonic ischemia as long as the marginal artery is
anterior surface, where there are few, if any, signifi- not disrupted. The kidney and surrounding tissues are
cant branches. dissected free from the posterior abdominal wall and
Superior to the renal vessels the peritoneum fans rotated medially, and the renal artery is identified,
out laterally, and the dissection is performed to the ligated, and divided. In bulky tumors the superior
lateral border of the peritoneum. In most larger tumors mesenteric artery might be displaced laterally; great
and some smaller tumors, the peritoneum cannot be care should be taken to distinguish the superior
dissected free of Gerota’s fascia, and the surgeon is mesenteric artery from the renal artery on either side.
forced to remove a window of peritoneum with the The ureter, gonadal vessels, and periureteral fat are
specimen. Care should be taken to avert injury to the dissected free of the posterior peritoneum and divided
bowel, especially the C portion of the duodenum. in two or three separate bundles. The dissection is then
The superior portion of the specimen, including the carried superiorly along the aorta on its anterior
adrenal gland, should be dissected free of the retro- surface, where there are few significant branches.
peritoneum and liver. Because there may be branches The splenorenal ligament is identified, ligated, and
of the phrenic and other vessels at this point, the author divided to avert splenic injury during mobilization of
generally uses a series of large hemoclips, dividing the the kidney. The superior portion of the specimen
tissue below the clips to enhance hemostasis. should be dissected free of the retroperitoneum.
On the left side, the descending colon is retracted The specimen should be free at this point except for
medially, and the lateral reflection of the peritoneum the venous structures. The left adrenal vein drains into

Fig. 7. Kidney and surrounding tissues dissected free from the posterior abdominal wall and rotated medially. (From El-Galley
RES, Keane TE. Kidneys, ureters, and bladders. In: Wood WC, Skandalakis JE, editors. Anatomic basis of tumor surgery.
St. Louis: Quality Medical Publishing; 1999; with permission.)
R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065 1061

the renal vein and is ligated and divided. The back Satinsky clamp is applied to the cavotomy, and the
surface of the renal vein should be inspected carefully edges of the vena cava are approximated gently with
for any lumbar veins, which if present should be Allis clamps. The tourniquet on the contralateral renal
ligated and divided. Then the renal vein should be vein and infrarenal vena cava and the clamp on the
palpated for possible unsuspected thrombi, divided, porta hepatis are released, leaving the tourniquet on
and ligated. Large tumors on either side frequently the suprahepatic vena cava in place. The Satinsky
develop parasitizing vessels, which are abnormal in clamp is briefly vented to allow the air in the venal
structure and frequently can lead to troublesome cava to be evacuated; then the clamp is closed again,
bleeding if not ligated or clipped with great care. and the last tourniquet on the vein is released. The
affected renal vein is transected flush with the vena
Management of tumor extension in the vena cava cava. The entire cavotomy is then closed with a
continuous 5-0 polypropylene suture.
The presence of a solid mass in the vena cava
might represent tumor extension into the lumen, Lymphadenectomy
blood thrombus, or less commonly tumor invasion
of the vena cava wall. Tumor extension into the vena The prognosis of renal cell carcinoma is mostly
cava occurs in 4% to 10% of cases, and tumor-free affected by presence or absence of nodal metastasis.
survival equivalent to survival of stage II is achieved Because of the position of the kidney just inferior to
by complete removal of tumor extension in patients the cisterna chylae, tumor spread from the renal
without lymph node involvement [23]. Exploration of lymphatic vessels to the cisterna chylae and wide-
the vena cava is a major procedure, and a complete spread dissemination of the disease is common.
set of vascular instruments should be available. The Curative lymphadenectomy is not possible in most
extent of the tumor extension into the vena cava cases, and the value of lymphadenectomy is limited
should be delineated preoperatively to help in plan- to the diagnosis of lymph node involvement. Limited
ning the surgical approach. Right-sided renal tumors dissection of the tissue around the junction of the
with limited vena caval extension can be approached renal vessel to the nearest great vessel and resection
with a right flank incision. A thoracoabdominal of the visible or palpable nodes is usually sufficient.
incision is used for high right-sided tumor extension,
whereas a midline incision with or without a median Nephroureterectomy
sternotomy extension is frequently required for
patients with left renal tumors and vena caval exten- Transitional cell carcinoma of the calyces, pelvis,
sion to the level of the hepatic veins or above. or ureter usually is treated with nephroureterectomy,
Exposure of the retrohepatic vena cava is started provided the contralateral collecting system is normal
with division of the right triangular and coronary and no evidence exists of distant metastasis. Pre-
ligaments of the liver and ligation of the small hepatic operative evaluation should include cystoscopy and
(caudate) veins. The liver is then mobilized medially bilateral retrograde pyelography for better evaluation
to expose the vena cava, and a cardiac tourniquet is of the collecting system. The operation can be per-
applied around the vessel for temporary occlusion. formed through a flank incision with downward
The contralateral renal vein and the infrarenal vena extension, or alternatively two separate incisions or
cava also are occluded with a Rumel tourniquet. a midline incision can be made. The technique of
Because about one fourth of the venous return in nephrectomy is the same. The ureter is mobilized
the vena cava comes from the liver, clamping the with blunt and sharp dissection down to its insertion
porta hepatis through the foramen of Winslow with a in the bladder. A cuff of the bladder must be removed
noncrushing vascular clamp reduces the blood loss with the lower ureter because this is the most com-
remarkably. A cavotomy is made adjacent to the mon site for tumor recurrence after nephroureterec-
hepatic veins and extended inferiorly to the origin tomy. The bladder is then closed in two layers with
of the affected renal vein. A 20F Foley catheter with a 2-0 chromic catgut sutures. A Foley catheter is left in
30-mL balloon is introduced into the vena cava, and the bladder for drainage, and a drain in the pelvis
the balloon is inflated above the level of the thrombus next to the suture line.
and withdrawn gently to extract the thrombus out of
the vena cava. In the rare occasion when the tumor Partial nephrectomy
invades the wall of the vein, partial or complete
resection of the vein is considered. Air should be Renal cell carcinoma in a solitary functioning
evacuated from the vena cava before closure. A kidney or bilateral tumors is best treated with partial
1062 R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065

nephrectomy. Full preoperative evaluation should be lymphadenopathy, IVC involvement, and extensive
performed to confirm that the disease is localized. perinephric visceral involvement. Unlike laparoscopic
The arterial anatomy of the affected kidney should be radical nephrectomy, the laparoscopic technique for
studied carefully with preoperative angiography. partial nephrectomy is being developed. The increased
Flank incisions through the bed of the eleventh or popularity of hand-assisted laparoscopy in the past few
twelfth rib, with attempt to stay extrapleural and years has reduced the learning curve for many sur-
extraperitoneal, provide an excellent exposure of the geons and enabled the surgeons to deal with large
peripheral renal vessels. Then the kidney is mobilized complicated tumors in safe and effective manner
within Gerota’s fascia. Temporary occlusion of the [25,26].
renal artery and surface cooling of the kidney with Full laparoscopic radical nephrectomy is per-
iced slush during the procedure allow 60 minutes of formed through three to four laparoscopic ports and
operating time without significant ischemic injury to hand-assisted laparoscopy is performed through a
the kidney. For longer procedures the kidney should hand port incision (6 to 8 cm) and two to three
be perfused with cold Collin’s solution through an instrument ports. The positions for these ports are
arterial catheter, which allows 3 hours for surgery. variable according the surgeon’s preference. The
Small polar or peripheral renal tumors may not abdomen is insufflated with CO2 to a pressure of
require renal artery occlusion, however, and the seg- 25 mm hg. The peritoneum is incised at the Toldt’s
mental artery instead can be identified and divided. line to mobilize the colon and expose the Gerota’s
Simple enucleation for malignant lesions should be fascia, which should be kept intact. On the right side,
avoided even if the tumor looks well defined, because the duodenum is mobilized medially and the liver is
of the probable presence of microscopic extensions of retracted. The renal pedicle is then dissected and the
these tumors beyond the pseudocapsule. Tumors of the ureter is divided. The artery and vein are individually
upper or lower pole of the kidney are best resected by clamped and divided with a laparoscopic stapler. On
polar nephrectomy (guillotine resection), whereas the left side, the spleen and tail of the pancreas should
mid-renal tumors are resected with wedge resection. be mobilized medially by dividing the splenorenal
and splenophrenic ligaments. Then, the kidney is
Laparoscopic surgery dissected as on the right side. The kidney is extracted
through the hand port incision if hand-assisted lapa-
Over the past few years, laparoscopic surgery has roscopy is chosen. If full laparoscopy is used, a
become more popular than the standard open radical separate incision in the suprapubic area is made for
nephrectomy in many centers and is rapidly becoming kidney extraction. Adrenalectomy used to be part of
the standard of care in most patients with stage T1 and radical nephrectomy in most patients. More recent
T2 tumors (Fig. 8). Laparoscopic surgery offers data, however, showed that adrenalectomy is not
smaller cosmetic scars, better visualization, less blood necessary for lower pole tumors [27 – 29].
loss, minimal trauma, and equivalent tumor control to Laparoscopic partial nephrectomy has been a
open surgery, which is reflected in reduced morbidity difficult procedure even for the experienced laparos-
and increased patient satisfaction [24]. The contra- copist. The laparoscopic approach is similar to radical
indications to laparoscopic surgery include bulky nephrectomy. After the kidney is dissected, the tumor
is located, preferably with the aid of intraoperative
ultrasound using a laparoscopic probe. The tumor is
then excised with a safety margin. Frozen sections are
obtained to ensure complete resection and hemostasis
is secured with electrocautery, fibrin glue, or sutures.
If the collecting system is opened, it should be closed
with water-tight sutures. The most difficult part of this
procedure is obtaining hemostasis. Desai et al [30]
have suggested a technique similar to open partial
nephrectomy. The kidney is dissected, the renal vessels
are dissected and clamped with laparoscopic Pull Dog
clamps, and the kidney is cooled with ice slush before
excising the tumor area.
An alternative to partial nephrectomy for small
renal tumors is the destruction of the tumor area with
Fig. 8. Laparoscopic view during radical nephrectomy. freezing or radiofrequency ablation. These techniques
R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065 1063

may be performed percutaneously under CT guidance lymph node metastasis is 17% and 5%, respectively
or laparoscopically. Renal tumor ablation is discussed [41]. Minervini et al [42] compared the survival for
elsewhere in this issue. renal cell carcinoma patients who had regional lym-
phadenectomy compared with patients who were
managed by radical nephrectomy alone. The 5-year
Results overall survival was similar whether or not lympha-
denectomy was performed: 79% for radical nephrec-
Local disease control for localized renal tumors is tomy alone and 78% for radical nephrectomy and
currently achieved in most patients who were treated regional lymphadenectomy.
with radical or partial nephrectomy. If the tumor is Since the introduction of laparoscopic radical and
completely removed with negative margins, local total nephrectomy for renal tumor in June 1990, it has
recurrence is rare. In most series local recurrence is been applied successfully worldwide to hundreds of
reported to occur in less than 2% of patients after patients [43]. Recent data have shown this procedure
radical nephrectomy [31]. Similarly, in a large series to produce cancer control identical to that of open
from Cleveland Clinic, local recurrence was reported radical and total nephrectomy [44]. Although in most
in 3.2% of patients who were treated with partial centers the cost of the procedure remains higher than
nephrectomy; half of these patients had distant me- open surgery, the patient benefits of decreased pain,
tastasis in addition to local recurrence [32,33]. The reduced hospitalization, less blood loss, and more
cancer-specific survival for patients who were treated rapid convalescence seem to be universal [45]. At this
with radical nephrectomy was estimated to be 86.6%, time, laparoscopic radical and total nephrectomy for
74%, 68.7%, and 63.8% for 1, 3, 7, and 10 years, the treatment of renal tumors should become the new
respectively [34]. Local recurrence in the absence of standard of care in many centers [46].
metastasis is usually amenable to local excision.
Schrodter et al [35] reported on 16 patients who were
diagnosed with local recurrence after radical nephrec-
tomy. At surgical exploration, three patients were References
found to have a false-positive CT, whereas the others
had true local recurrence of their tumors. After [1] McLaughlin JK, Lipworth L. Epidemiologic aspects of
renal cell cancer. Semin Oncol 2000;27:115 – 23.
complete resection of the recurrence, seven patients
[2] Lee CT, Katz J, Fearn PA, Russo P. Mode of presenta-
died of metastatic disease after a mean survival of tion of renal cell carcinoma provides prognostic infor-
23 months, and six patients were alive with a mean mation. Urol Oncol 2002;7:135 – 40.
follow-up of 53 months [35]. [3] Bennouna J, Delva R, Gomez F, Lesimple T, Geoffrois
Involvement of the IVC with tumor thrombi occurs L, Linassier C, et al. A phase II study with 5-fluoro-
in 4% to 10% of patients. Tumor excision is possible in uracil, folinic acid and oxaliplatin (FOLFOX-4 regi-
most of these patients with a survival range of 77% at men) in patients with metastatic renal cell carcinoma.
2 years and 55% at 5 years for patients who did have Oncology 2003;64:25 – 7.
other evidence of metastasis [36]. Bissada et al [37] [4] Escudier B, Lassau N, Couanet D, Angevin E, Mesrati
reported on the outcome of the management of renal F, Leborgne S, et al. Phase II trial of thalidomide in
renal-cell carcinoma. Ann Oncol 2002;13:1029 – 35.
cell carcinoma with IVC extension in 54 patients.
[5] Ryan CW, Vogelzang NJ, Stadler WM. A phase II trial
Forty-eight patients did not have evidence of other of intravenous gemcitabine and 5-fluorouracil with
metastasis. Seven patients had invasion of the IVC subcutaneous interleukin-2 and interferon-alpha in pa-
wall and required partial or complete resection of the tients with metastatic renal cell carcinoma. Cancer
IVC. Of the 48 patients without evidence of metastasis 2002;94:2602 – 9.
at surgery, the perioperative mortality rate was 2%. [6] Gilbert SM, Russo P, Benson MC, Olsson CA,
Twenty-two patients (47%) were alive without evi- McKiernan JM. The evolving role of partial nephrec-
dence of metastases, 4% developed solitary metastasis, tomy in the management of renal cell carcinoma.
and 36% eventually developed multiple metastases. Curr Oncol Rep 2003;5:239 – 44.
The follow-up ranged from 25 to 144 months. [7] Guillonneau B, Bermudez H, Gholami S, El Fettouh H,
Gupta R, Adorno Rosa J, et al. Laparoscopic partial
It is generally accepted that patients with lymph
nephrectomy for renal tumor: single center experience
node metastasis have a poor prognosis regardless of comparing clamping and no clamping techniques of
the completeness of lymphadenectomy [38,39]. The the renal vasculature. J Urol 2003;169:483 – 6.
reported incidence of lymph node metastasis in renal [8] Adkins KL, Chang SS, Cookson MS, Smith Jr JA. Par-
cancer patients is 13% [40]. The 5- and 10-year tial nephrectomy safely preserves renal function in pa-
survival for patients with renal cell carcinoma and tients with a solitary kidney. J Urol 2003;169:79 – 81.
1064 R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065

[9] Flanigan RC, Yonover PM. The role of radical ne- [27] Moudouni SM, En-Nia I, Patard JJ, Manunta A, Guille
phrectomy in metastatic renal cell carcinoma. Semin F, Lobel B. Real indications for adrenalectomy in renal
Urol Oncol 2001;19:98 – 102. cell carcinoma. Scand J Urol Nephrol 2002;36:273 – 7.
[10] Anton P, Kirchner H, Jonas U, Atzpodien J. Cytokines [28] Paul R, Mordhorst J, Busch R, Leyh H, Hartung R.
and tumor vaccination. Cancer Biother Radiopharm Adrenal sparing surgery during radical nephrectomy in
1996;11:315 – 8. patients with renal cell cancer: a new algorithm. J Urol
[11] Jeon SH, Chang SG, Kim JI. The role of adjuvant 2001;166:59 – 62.
immunotherapy after radical nephrectomy and prog- [29] Tsui KH, Shvarts O, Barbaric Z, Figlin R, de Kernion
nostic factors in pT3N0M0 renal cell carcinoma. Anti- JB, Belldegrun A. Is adrenalectomy a necessary com-
cancer Res 1999;19:5593 – 7. ponent of radical nephrectomy? UCLA experience
[12] Sawczuk IS, Pollard JC. Renal cell carcinoma: should with 511 radical nephrectomies. J Urol 2000;163:
radical nephrectomy be performed in the presence of 437 – 41.
metastatic disease? Curr Opin Urol 1999;9:377 – 81. [30] Desai MM, Gill IS, Kaouk JH, Matin SF, Novick AC.
[13] Auberton E, Bellin MF, Richard F, Chatelain C, Del- Laparoscopic partial nephrectomy with suture repair of
court A, Grellet J. Comparative study of MRI and CT the pelvicaliceal system. Urology 2003;61:99 – 104.
x-ray in evaluating the extension of kidney cancer in [31] Itano NB, Blute ML, Spotts B, Zincke H. Outcome of
adults. J Radiol 1989;70:327 – 36. isolated renal cell carcinoma fossa recurrence after
[14] Remer EM, Herts BR, Veniero JC. Imaging for nephron- nephrectomy. J Urol 2000;164:322 – 5.
sparing surgery. Semin Urol Oncol 2002;20:180 – 91. [32] Hafez KS, Fergany AF, Novick AC. Nephron sparing
[15] Mazer MJ, Quaife MA. Hypertrophied column of Ber- surgery for localized renal cell carcinoma: impact of
tin pseudotumor: radionuclide investigation. Urology tumor size on patient survival, tumor recurrence and
1979;14:210 – 1. TNM staging. J Urol 1999;162:1930 – 3.
[16] Thornbury JR, McCormick TL, Silver TM. Anatomic/ [33] Novick AC. Nephron-sparing surgery for renal cell
radiologic classification of renal cortical nodules. AJR carcinoma. Annu Rev Med 2002;53:393 – 407.
Am J Roentgenol 1980;134:1 – 7. [34] Frank I, Blute ML, Cheville JC, Lohse CM, Weaver AL,
[17] Bosniak MA. The current radiological approach to re- Zincke H. An outcome prediction model for patients
nal cysts. Radiology 1986;158:1 – 10. with clear cell renal cell carcinoma treated with radical
[18] Bosniak MA. Difficulties in classifying cystic lesions nephrectomy based on tumor stage, size, grade and ne-
of the kidney. Urol Radiol 1991;13:91 – 3. crosis: the SSIGN score. J Urol 2002;168:2395 – 400.
[19] Bosniak MA. How does one deal with a renal cyst that [35] Schrodter S, Hakenberg OW, Manseck A, Leike S,
appears to be Bosniak class II on a CT scan but that has Wirth MP. Outcome of surgical treatment of isolated
sonographic features suggestive of malignancy (e.g., local recurrence after radical nephrectomy for renal
nodularity of wall or a nodular, irregular septum)? cell carcinoma. J Urol 2002;167:1630 – 3.
AJR Am J Roentgenol 1994;163:216. [36] Babu SC, Mianoni T, Shah PM, Goyal A, Choudhury
[20] Koga S, Nishikido M, Inuzuka S, Sakamoto I, Hayashi M, Eshghi M, et al. Malignant renal tumor with exten-
T, Hayashi K, et al. An evaluation of Bosniak’s radio- sion to the inferior vena cava. Am J Surg 1998;176:
logical classification of cystic renal masses. BJU Int 137 – 9.
2000;86:607 – 9. [37] Bissada NK, Yakout HH, Babanouri A, Elsalamony T,
[21] Bosniak MA. The use of the Bosniak classification sys- Fahmy W, Gunham M, et al. Long-term experience
tem for renal cysts and cystic tumors. J Urol 1997;157: with management of renal cell carcinoma involving
1852 – 3. the inferior vena cava. Urology 2003;61:89 – 92.
[22] Schatz SM, Lieber MM. Update on oncocytoma. Curr [38] Kitamura Y, Watanabe M, Komatsubara S, Sakata Y.
Urol Rep 2003;4:30 – 5. Renal cell carcinoma with regional lymph node metas-
[23] Kaplan S, Ekici S, Dogan R, Demircin M, Ozen H, tasis. Hinyokika Kiyo 1995;41:433 – 8.
Pasaoglu I. Surgical management of renal cell carcino- [39] Marshall FF. Radical nephrectomy. Urology 1995;46:
ma with inferior vena cava tumor thrombus. Am J Surg 153 – 4.
2002;183:292 – 9. [40] Terrone C, Guercio S, De Luca S, Poggio M, Castelli
[24] Portis AJ, Yan Y, Landman J, Chen C, Barrett PH, Fentie E, Scoffone C, et al. The number of lymph nodes
DD, et al. Long-term followup after laparoscopic radical examined and staging accuracy in renal cell carcinoma.
nephrectomy. J Urol 2002;167:1257 – 62. BJU Int 2003;91:37 – 40.
[25] Landman J, Lev RY, Bhayani S, Alberts G, Rehman J, [41] Golimbu M, Joshi P, Sperber A, Tessler A, Al-Askari
Pattaras JG, et al. Comparison of hand assisted and S, Tessler A, Morales P. Renal cell carcinoma: survival
standard laparoscopic radical nephroureterectomy for and prognostic factors. Urology 1986;27:291 – 301.
the management of localized transitional cell carcino- [42] Minervini A, Lilas L, Morelli G, Traversi C, Battaglia
ma. J Urol 2002;167:2387 – 91. S, Cristofani R, et al. Regional lymph node dissection
[26] Nelson CP, Wolf Jr JS. Comparison of hand assisted in the treatment of renal cell carcinoma: is it useful in
versus standard laparoscopic radical nephrectomy for patients with no suspected adenopathy before or during
suspected renal cell carcinoma. J Urol 2002;167: surgery? BJU Int 2001;88:169 – 72.
1989 – 94. [43] Clayman RV, Kavoussi LR, Soper NJ, Dierks SM,
R. El-Galley / Radiol Clin N Am 41 (2003) 1053–1065 1065

Clayman RV, Darcy MD, et al. Laparoscopic nephrec- Heidorn C, McDougall EM, et al. Laparoscopic versus
tomy. N Engl J Med 1991;324:1370 – 1. open radical nephrectomy: a 9-year experience. J Urol
[44] El Fettouh HA, Rassweiler JJ, Schulze M, Salomon L, 2000;164:1153 – 9.
Allan J, Ramakumar S, et al. Laparoscopic radical [46] Portis AJ, Clayman RV. Should laparoscopy be the
nephroureterectomy: results of an international multi- standard approach used for radical nephrectomy? Curr
center study. Eur Urol 2002;42:447 – 52. Urol Rep 2001;2:165 – 70.
[45] Dunn MD, Portis AJ, Shalhav AL, Elbahnasy AM,
Radiol Clin N Am 41 (2003) 1067 – 1075

Percutaneous image-guided radiofrequency ablation


of renal malignancies
Ronald J. Zagoria, MD
Department of Radiology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem,
NC 28401 – 1088, USA

The treatment of renal cell carcinoma (RCC) can sizeable portal to introduce the ablation device.
be a vexing problem. Although advanced disease Cryotherapy usually requires laparoscopic or open
does not respond well to therapy and the prognosis surgery. Although radiofrequency devices can be
remains extremely poor, the rate of RCC diagnosis introduced intraoperatively during an open procedure,
has increased substantially [1]. In the year 2002, there most experience with this technique has used percu-
were over 30,000 new cases of RCC diagnosed in the taneous image-guided procedures.
United States [2]. This equates to greater than a 100% Radiofrequency ablation uses the introduction of a
increase in the incidence of RCC diagnosed in the high-frequency, alternating current within the targeted
United States since 1950. Most of this increase has tissue [7]. Emission of this energy in a patient to
occurred because of the diagnosis of small, localized whom grounding pads have been applied results in
tumors detected incidentally in asymptomatic patients concentrated ionic agitation in tissues nearby the site
imaged for other reasons [3]. of energy transmission. This ionic agitation in turn
Radical nephrectomy has long been considered results in the generation of heat. This type of thermal
the standard treatment for localized RCC. Mean- ablative technique is akin to microwave heating used
while, renal-sparing surgery has grown in popularity in everyday applications. When living human tissues
and the techniques have been refined. Studies com- are heated above 49°C immediate cell death occurs
paring surgical techniques have shown that open [14]. The cell death is induced by denaturation of
partial nephrectomy is as effective in curing small, protein, melting of cell membranes, and thermal
localized RCCs as radical nephrectomy [4,5]. This destruction of cytoplasm [14]. This results in direct
indicates that complete eradication of a renal tumor cytodestruction of the affected cells. Some cells are
can result in cure rates comparable with those of destroyed at temperatures below 49°C, but some cells
treatment using complete removal of a kidney con- can survive temperatures approaching 49°C. For per-
taining a tumor. Advances in imaging and thermal cutaneous image-guided RFA the energy is delivered
ablation techniques, combined with the theory that into the target tissue through needle-shaped probes.
tumor destruction yields results comparable with Currently available RFA electrodes range in diameter
tumor resection, have led to increased interest in from 15 to 17 gauge. Three radiofrequency devices
image-guided, minimally invasive percutaneous ther- approved by the Food and Drug Administration are
mal ablative techniques for the treatment of RCC. available in the United States. Each of these uses a
There is substantial experience in treatment of neo- different strategy to maximize the size of thermal
plasms using both radiofrequency ablation (RFA) ablation. Each device also uses a slightly different
[6 – 9], which causes tumor destruction by heating, approach to energy delivery for thermal destruction.
and cryotherapy, which destroys tumors using freez- All of the available radiofrequency devices use genera-
ing [10 – 13]. Most cryotherapy devices require a tors that deliver between 150 and 200 W of energy.
This represents an increase over earlier generators that
delivered 50 to 125 W of energy. This lower energy
E-mail address: rzagoria@wfubmc.edu proved suboptimal. The next generation of radio-

0033-8389/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0033-8389(03)00074-5
1068 R.J. Zagoria / Radiol Clin N Am 41 (2003) 1067–1075

frequency generators may deliver even higher levels of zone is increased by inducing ischemia, or in devas-
energy to increase the area of the treatment zone. cularized tissue. Alternatively flowing blood, large
The maximum size of the treatment zone for in fluid-containing spaces, or circulating air can de-
vivo treatment of renal tumors has been shown to crease the effective size of the treatment zone [14].
approximate a 5 cm sphere for a single RFA [7]. The When RFA of renal tumors is performed using a
size of the zone of ablation is often smaller than this percutaneous technique, imaging guidance is required.
maximum, and usually averages between a 3- and This is usually done using CT scanning or sonography.
4-cm sphere [14]. The maximum size of the treatment The technique of placing the electrode is analogous to
R.J. Zagoria / Radiol Clin N Am 41 (2003) 1067–1075 1069

that of performing an image-guided biopsy of a renal identified in the necrotic regions. Evaluation of the
mass (Fig. 1). The actual treatment of the tumor can be surrounding kidney following nephrectomy demon-
quite painful; more sedation is required than for a strated that a margin of treated, nonviable kidney tissue
standard needle biopsy. Most cases are performed with was identified in all eight of the completely ablated
conscious sedation and local anesthesia [7]. tumors. The treatment margin ranged from 2 to 13 mm
Percutaneous RFA has been applied for the pri- in thickness. In no case was the rim of destroyed
mary treatment of RCC in nonoperative patients normal kidney greater than 13 mm in diameter.
[6,8,9], for the treatment of local recurrences of The results of this study are encouraging and indi-
RCC that are deemed inoperable [15], and also for cate that in vivo treatment of small RCCs is feasible,
the treatment of isolated metastases from RCC although clearly some tumors require greater than
primaries [1]. At the author’s institution they per- one 12-minute RFA treatment for complete destruc-
formed a phase two clinical trial using the in vivo tion of the tumor. Based on this study it seems that
application of RFA of renal parenchymal tumors only a small amount of adjacent normal kidney is
immediately before nephrectomy [7]. Biopsies were destroyed, suggesting that little diminution in renal
obtained of all tumors before RFA. The ablation was function results when using this technique.
performed intraoperatively using ultrasound guidance. In 1992, the first published report using radio-
In this study, a Radionics cool-tip radiofrequency frequency tissue ablation was released. This used
system (Radionics, Burlington, MA), which consists RFA for the treatment of hepatic tissues [16]. In
of the CC-1 Cosman Coagulator, the cool-tip treatment 1997, the use of RFA to produce extensive necrosis
electrode, and a peristaltic perfusion pump, was used. of kidney tumors in humans was reported [17]. In
Following tumor biopsy a single RFA of 12 minutes 1998, a case report was published reporting the first
duration was performed using the automatic output case where percutaneous RFA was used for the
control setting of this radiofrequency generator. Fol- complete treatment of an RCC in a human under
lowing the single RFA a nephrectomy was performed ultrasound guidance [18].
and the tumor was evaluated histologically using both Since those reports were published, several other
standard and vital stains. larger studies using RFA for the treatment of renal
The tumors treated in this study were all deter- tumors have been published [6 – 9]. One group has
mined to be RCCs (nine clear cell carcinomas and used RFA ablation in a porcine model and found that
one papillary carcinoma). The tumors ranged in size RFA of renal tumors results in necrosis of the ablated
from 1.4 to 8 cm in diameter. The average tumor size tumor and surrounding renal parenchyma with no
in this series was 3.2 cm in diameter. Eight of the evidence of collecting system damage [19]. One
10 tumors were completely destroyed with no identi- group has reported their experience treating nine
fiable viable tumor remaining after a single 12-minute renal tumors in eight patients using percutaneous
ablation. Two tumors, 1.4 and 8 cm in diameter, RFA [6]. With a follow-up of just over 10 months
were incompletely treated with a single ablation. The in these eight patients, seven of the nine tumors were
1.4-cm tumor had approximately 70% destruction but completely free of demonstrable enhancement on CT
the temperature in the tissue immediately following the scans suggesting complete ablation of these seven
ablation was below the target temperature of 49°C or tumors. Within this group of tumors, five of five
higher. A large segment of the 8-cm tumor was exophytic tumors were rendered completely non-
destroyed with viable tumor remaining at the periph- enhancing, whereas only one of three central RCCs
ery. In all cases no skip areas of viable tumor were was completely free of enhancement on follow-up

Fig. 1. The technique of radiofrequency ablation (RFA) of renal malignancies in an elderly man with multiple co-morbidities and
an enlarging biopsy-proved renal cell carcinoma (RCC). (A) Unenhanced CT shows the 3-cm RCC (arrow) extending from the
upper pole of the right kidney. This measured 22 HU on this scan. (B) Following contrast material injection this tumor enhanced
to 99 HU on this scan obtained several months before the ablation procedure. (C) CT scan obtained during the RFA procedure
shows the patient in a prone position with the percutaneous radiofrequency electrode (arrow) placed so the tip is bisecting the
tumor. There is a small amount of blood (arrowhead) in the perinephric space, a common finding seen during this procedure. A
22-gauge needle is seen adjacent to the electrode. This was used to target the tumor using a tandem technique. (D) An
unenhanced CT scan obtained 14 months after the ablation shows the ablated RCC is slightly hyperdense compared with the
kidney. It measured 33 HU on this scan. There is a small amount of perinephric stranding seen adjacent to the kidney, an
expected finding following percutaneous RFA. (E) Following contrast material injection the mass shows no enhancement
measuring 35 HU. Lack of enhancement and stability of the tumor size strongly suggests complete tumor destruction.
1070 R.J. Zagoria / Radiol Clin N Am 41 (2003) 1067–1075

CT. All three RCCs smaller than 3 cm were rendered particularly those located centrally. Although micro-
completely nonenhancing, whereas four of six RCCs scopic hematuria was common in the 24-hour period
greater than 3 cm were completely ablated. These immediately following ablation there were no serious
authors reported that multiple ablations within a complications in any of these eight patients.
single treatment session, or repeated treatment ses- In one larger series 24 RCCs in 21 patients with
sions, are often needed to destroy RCCs completely, von Hippel-Lindau disease or with familial papillary

Fig. 2. MR imaging of an RCC before and after percutaneous RFA showing residual viable tumor requiring repeat ablation.
(A) T1-weighted MR image before gadolinium injection shows the 3-cm RCC arising from the upper pole of the right kidney.
(B) T1-weighted MR image at the same level following gadolinium injection shows marked heterogeneous enhancement of the
tumor. (C) CT scan obtained during the RFA procedure shows the patient in a prone position with the percutaneous
radiofrequency electrode (arrow) placed so the tip is bisecting the tumor in the right kidney. There is a small amount of blood
(arrowhead) in the perinephric space from this procedure. (D) T1-weighted MR image before gadolinium injection obtained
4 months after the percutaneous renal tumor ablation shows heterogeneous increased signal in much of the tumor. This is
commonly seen and is believed caused by coagulative necrosis in the ablated tumor. (E) Following gadolinium injection this
T1-weighted MR image with fat saturation shows a small area of enhancement (arrows) in the anterior aspect of the otherwise
nonenhancing tumor. This enhancement was interpreted as an area of residual viable tumor. The perinephric hemorrhage that
occurred during the ablation is again seen. ( F) CT scan obtained during the second RFA procedure shows the patient in a prone
position with the percutaneous radiofrequency electrode (arrow) placed so the tip is located in the area where viable tumor was
demonstrated on the MR image shown in Fig. 2E. ( G) This T1-weighted MR image with fat saturation and following gadolinium
injection was obtained 4 months after the second percutaneous ablation. No enhancement was detectable within the treated tumor
suggesting complete tumor destruction.
R.J. Zagoria / Radiol Clin N Am 41 (2003) 1067–1075 1071

RCCs were treated with percutaneous RFA [8]. At a In many cases coagulative necrosis within the
2-month follow-up, 19 of the 24 treated tumors treated area often has a higher baseline attenuation
demonstrated no evidence of contrast enhancement and high signal on T1 and T2 sequences when
on CT imaging. The remaining five tumors demon- imaged with CT or MR imaging, respectively
strated some persistent enhancement. There were no (see Fig. 1; Fig. 2). This should not be misinterpreted
serious complications in this series of patients and all as viable neoplasm. Areas of enhancement should be
of these patients were treated on an outpatient basis. viewed, however, as residual viable tumor and re-
Contrast-infused CT and MR imaging have been treated (see Fig. 2).
used to detect viable tumor following renal tumor There is a growing body of knowledge regarding
RFA. In one in vivo study there were no skip areas percutaneous RFA of RCCs. It seems this technique
where viable tumor survived within the ablation zone has a low complication rate, preserves renal function,
[7], so these should be reliable methods for following is well tolerated by patients, and can result in com-
these patients. In a second in vivo study using a plete destruction of renal tumors 5 cm or smaller in
different RFA device, however, 5% to 10% viable most patients (Fig. 3). In some patients viable tumor
tumor remained in most tumors [20]. This group also can be demonstrated following one session of RFA
found that contrast enhancement could not always be (see Fig. 2). This may require further ablation ses-
detected with CT in areas where viable tumor was sions (see Fig. 2). Repeated RFA ablations have been
histologically demonstrated following nephrectomy performed in many patients with renal tumors [6] and
[20]. This suggests the possibility that lack of contrast routinely in patients with hepatic tumors, where RFA
enhancement may overestimate tumor destruction experience is more extensive than in the kidney.
caused by RFA. Repeat RFA for treatment of residual tumor seems

Fig. 2 (continued ).
1072 R.J. Zagoria / Radiol Clin N Am 41 (2003) 1067–1075

Fig. 3. Successful ablation of a larger, centrally located RCC. (A) Unenhanced CT scan shows a 5 cm  3 cm tumor (arrow) located
centrally in the left kidney. (B) Contrast-enhanced CT obtained immediately following percutaneous ablation shows heterogeneity
of the treated tumor without enhancement. There is perinephric hemorrhage present resulting from the ablation procedure.

to be a safe procedure without a risk greater than with treatment was performed resulting in a marked de-
primary RFA. It seems that there is little renal damage crease in symptoms and improved quality of life. In
associated with RFA. Even in the treatment of central an additional case [15], recurrence of RCC in the
tumors the development of clinically important pel- surgical bed occurred (Fig. 4). The recurrence abutted
vicalyceal damage has not been reported. Because in the abdominal aorta and was believed to be unresect-
vivo studies have demonstrated only a small amount able (see Fig. 4). The recurrence was unresponsive to
of kidney destruction in the area surrounding the immunotherapy and chemotherapy and continued to
tumor, renal function should remain nearly intact grow during CT surveillance monitoring. A single
following this procedure. Obviously, there is limited session of percutaneous CT-guided RFA was per-
information on this procedure and at this time it formed on this recurrent tumor (see Fig. 4). An
should be reserved for patients for whom some immediate contrast-infused CT demonstrated no evi-
treatment is indicated, but who are not surgical dence of complications. No enhancing tumor could
candidates. Long-term results for this procedure have be demonstrated immediately following the RFA
yet to be reported. treatment. This patient has remained free of identifi-
Radiofrequency ablation also has been used for able disease (see Fig. 4) for over 16 months [15]. The
other applications in treating RCCs. A single case treated area of recurrence has decreased in volume
report described the successful use of RFA to treat with no evidence of enhancing, viable tumor. In these
intractable gross hematuria resulting from a large RCC two cases RFA seemed to be helpful in the treatment
[21]. In this case, the life-threatening hematuria failed of locally recurrent RCC. This is a very promising
to resolve following standard techniques including area for the use of image-guided RFA therapy.
renal embolization therapy. RFA was performed with- In addition, in some cases patients with distant
out complications and the patient remained free of metastases from RCC may experience prolonged
hematuria for a prolonged period of time. In this case, survival with eradication of the metastases [22]. This
RFA was used for palliation of symptoms from RCC well-documented phenomenon has most often been
rather than as a curative technique. seen in patients following metastasectomy of a small
Radiofrequency ablation also has been used for number of pulmonary metastases from RCC [22]. In
the treatment of recurrent and metastatic RCC. There general, patients with metastatic RCC have an ex-
have only been anecdotal successes reported; this tremely poor prognosis with nearly no 5-year sur-
technique must be viewed as preliminary, but prom- vivals following diagnosis [23]. Metastatic RCC is
ising. At the author’s institution they have used RFA nearly always incurable with systemic immuno-
to treat unresectable local recurrence from RCC in therapy or chemotherapy. Even though some case
two patients. In one patient the recurrence was reports show prolonged tumor remission following
extremely large and because of impingement on the metastasectomy, surgical resection of pulmonary
neural foramina was debilitating. Extensive RFA metastases uncommonly results in improved progno-
R.J. Zagoria / Radiol Clin N Am 41 (2003) 1067–1075 1073

Fig. 4. Percutaneous radiofrequency of a recurrent RCC. (A) Contrast-enhanced CT scan shows an enhancing mass (arrow)
abutting the aorta and the superior mesenteric artery in this man who previously had a left nephrectomy for RCC. A biopsy of
this mass was found to be recurrent RCC. (B) CT scan obtained during the RFA procedure shows the patient in a prone position
with the percutaneous radiofrequency electrode (arrow) placed so the tip is bisecting the tumor. This scan also shows a small
amount of gas in the tumor adjacent to the electrode. This common finding is attributable to necrosis and vaporization of tissue
induced by the radiofrequency energy. (C) Contrast-enhanced CT scan obtained 10 months after the ablation procedure shows
that tumor has decreased in size and no enhancement, consistent with complete tumor destruction. The superior mesenteric artery
and aorta remain widely patent.

sis for RCC patients [22]; surgeons are often reluc- this reported case is promising, but at this point it
tant to perform this procedure. This may be another should be reserved as a last resort in nonoperative
opportunity for the use of image-guided percutane- candidates until larger studies can demonstrate its
ous RFA treatment. In one published report a patient efficacy compared with surgery. Percutaneous RFA
with two pulmonary metastases from a previously has been used to treat neoplasms in sites where RCC
resected RCC was successfully treated with percuta- commonly metastasizes, however, including the
neous CT-guided RFA [24]. In this patient, the two lungs, liver, and the skeleton. The morbidity associ-
pulmonary metastases were peripherally located in ated with this type of treatment based on experience
the right lower lobe of the lung (Fig. 5). At 18-month with other neoplasms in these locations is very low
follow-up, this patient has not received any other with serious complications occurring in less than 5%
treatment for his RCC, but remains free of detectable of treated patients [25]. Because of the low morbidity
disease (see Fig. 5). The use of image-guided RFA and extremely low risk of mortality associated with
for treatment of isolated metastases from RCC as in image-guided percutaneous RFA, this treatment may
1074 R.J. Zagoria / Radiol Clin N Am 41 (2003) 1067–1075

Fig. 5. Percutaneous RFA of pulmonary metastasis from RCC. (A) CT shows one of two pulmonary metastases in this patient
with a history of RCC. These biopsy-proved metastases were refractory to systemic therapy. (B) CT scan obtained during the
RFA procedure shows the patient in a prone position with the percutaneous radiofrequency electrode placed so the tip is bisecting
the tumor in the right lung. (C) CT scan obtained 29 months after the ablation procedure shows a small area of scarring where the
tumor had been located. There was no evidence of viable tumor in either of the treated lung metastases.

be considered for patients who have failed systemic that is high, this must remain standard therapy for
therapy, and in whom the potential benefits of patients with potentially curable RCC. Some patients
surgery seem to be outweighed by the risk of with low-stage RCC, however, may not be surgical
substantial morbidity resulting from successful me- candidates. Image-guided RFA is an option for treat-
tastasis resection. ment of these patients. In addition, image-guided
RFA shows promise for the successful care of other
patients with RCC. In particular, RFA has been used
Summary successfully for the treatment of intractable hematuria
resulting from an RCC; local recurrences of RCC,
There is a growing body of experience supporting both for attempted cure and for palliation of symp-
the use of image-guided RFA for the treatment of toms; and finally for the treatment of isolated metas-
primary RCC. Because surgical resection is a tech- tases from RCC. As with the treatment of primary
nique with low mortality, and a proved success rate RCC, the data remain limited for these applications.
R.J. Zagoria / Radiol Clin N Am 41 (2003) 1067–1075 1075

This technique should be reserved until after standard [13] Shingleton WB, Sewell Jr PE. Percutaneous renal tu-
therapies have been exhausted. It seems likely that mor cryoablation with magnetic resonance imaging
some form of image-guided percutaneous tumor guidance. J Urol 2001;165:773 – 6.
[14] Goldberg SN, Gazelle GS, Mueller PR. Thermal abla-
therapy, such as RFA, will become an alternative
tion therapy for focal malignancy: a unified approach
treatment modality in some patients with potentially
to underlying principles, techniques, and diagnostic
curable RCC. imaging guidance. AJR Am J Roentgenol 2000;174:
323 – 31.
[15] McLaughlin CA, Chen MY, Torti FM, Hall MC, Zagoria
References RJ. Radiofrequency ablation of isolated local recurrence
of renal cell carcinoma after radical nephrectomy. AJR
[1] Zagoria RJ. Imaging of small renal masses: a medi- Am J Roentgenol 2003;181:93 – 4.
cal success story. AJR Am J Roentgenol 2000;175: [16] McGahan JP, Brock JN, Tessluk H, Gu WZ, Schneider
945 – 55. P, Browning PD. Hepatic ablation with use of radio-
[2] American Cancer Society. Cancer facts and figures, frequency electrocautery in the animal model. J Vasc
1996. Atlanta: American Cancer Society; 1996. Interv Radiol 1992;3:291 – 7.
[3] Rodriguez-Rubio FI, Diez-Caballero F, Martin-Mar- [17] Zlotta AR, Wildschutz T, Raviv G, et al. Radiofre-
quina A, Abad JI, Berian JM. Incidentally detected quency interstitial tumor ablation (RITA) is a possible
renal cell carcinoma. Br J Urol 1996;78:29 – 32. new modality for treatment of renal cancer: ex vivo
[4] Fergany AF, Hafez KS, Novick AC. Long-term results and in vivo experience. J Endourol 1997;11:251 – 8.
of nephron sparing surgery for localized renal cell car- [18] McGovern FJ, Wood BJ, Goldberg SN, Mueller PR.
cinoma: 10-year followup. J Urol 2000;163:442 – 5. Radio frequency ablation of renal cell carcinoma via
[5] Uzzo RG, Novick AC. Nephron sparing surgery for image guided needle electrodes. J Urol 1999;161:
renal tumors: indications, techniques and outcomes. 599 – 600.
J Urol 2001;166:6 – 18. [19] Rendon RA, Gertner MR, Sherar MD, et al. Develop-
[6] Gervais DA, McGovern FJ, Wood BJ, et al. Radio- ment of a radiofrequency based thermal therapy tech-
frequency ablation of renal cell carcinoma: early clini- nique in an in vivo porcine model for the treatment of
cal experience. Radiology 2000;217:665 – 72. small renal masses. J Urol 2001;166:292 – 8.
[7] Matlaga BR, Zagoria RJ, Woodruff RD, et al. Phase II [20] Rendon RA, Kachura JR, Sweet JM, et al. The un-
trial of radio frequency ablation of renal cancer: certainty of radio frequency treatment of renal cell
evaluation of the kill zone. J Urol 2002;168:2401 – 5. carcinoma: findings at immediate and delayed nephrec-
[8] Pavlovich CP, McClellan MW, Choyke PL, et al. Per- tomy. J Urol 2002;167:1587 – 92.
cutaneous radio frequency ablation of small renal tu- [21] Wood BJ, Grippo J, Pavlovich CP. Percutaneous radio
mors: initial results. J Urol 2002;167:10 – 5. frequency ablation for hematuria. J Urol 2001;166:
[9] Walther MM, Shawker TH, Libutti SK, et al. A phase 2 2303 – 4.
study of radio frequency interstitial tissue ablation of [22] Friedel G, Hurtgen M, Penzenstadler M, et al. Resec-
localized renal tumors. J Urol 2000;163:1424 – 7. tion of pulmonary metastases from renal cell carcino-
[10] Gill IS, Novick AC, Meraney AM, et al. Laparoscopic ma. Anticancer Res 1999;19:1593 – 6.
renal cryoablation in 32 patients. Urology 2000;56: [23] Figlin RA. Renal cell carcinoma: management of ad-
748 – 53. vanced disease. J Urol 1999;161:381 – 7.
[11] Rukstalis DB, Khorsandi M, Garcia FU, et al. Clinical [24] Zagoria RJ, Chen MY, Kavanagh PV, et al. Radio fre-
experience with open renal cryoablation. Urology 2001; quency ablation of lung metastases from renal cell car-
57:34 – 9. cinoma. J Urol 2001;166:1827 – 8.
[12] Shingleton WB, Farabaugh P, Hughson M, et al. Per- [25] Zagoria RJ, Chen MY, Shen P, et al. Complications
cutaneous cryoablation of porcine kidneys with mag- from radiofrequency ablation of liver metastases. Am
netic resonance imaging monitoring. J Urol 2001;166: Surg 2002;68:204 – 9.
289 – 91.

You might also like