You are on page 1of 14

European Journal of Radiology 141 (2021) 109777

Contents lists available at ScienceDirect

European Journal of Radiology


journal homepage: www.elsevier.com/locate/ejrad

Review

The role of imaging in the management of renal masses


Athina C. Tsili a, *, Efthimios Andriotis b, Myrsini G. Gkeli c, Miltiadis Krokidis d, e,
Myrsini Stasinopoulou b, Ioannis M. Varkarakis f, Lia-Angela Moulopoulos d, On behalf of the
Oncologic Imaging Subcommittee Working Group of the Hellenic Radiological Society
a
Department of Clinical Radiology, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45110, Ioannina, Greece
b
Department of Newer Imaging Methods of Tomography, General Anti-Cancer Hospital Agios Savvas, 11522, Athens, Greece
c
1st Department of Radiology, General Anti-Cancer Hospital Agios Savvas, 11522, Athens, Greece
d
1st Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, Areteion Hospital, 11528, Athens, Greece
e
Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern University Hospital, University of Bern, 3010, Bern, Switzerland
f
2nd Department of Urology, National and Kapodistrian University of Athens, Sismanoglio Hospital, 15126, Athens, Greece

A R T I C L E I N F O A B S T R A C T

Keywords: The wide availability of cross-sectional imaging is responsible for the increased detection of small, usually
Renal neoplasms asymptomatic renal masses. More than 50 % of renal cell carcinomas (RCCs) represent incidental findings on
Renal cell carcinoma noninvasive imaging. Multimodality imaging, including conventional US, contrast-enhanced US (CEUS), CT and
Multimodal imaging
multiparametric MRI (mpMRI) is pivotal in diagnosing and characterizing a renal mass, but also provides in­
Minimally invasive treatment
formation regarding its prognosis, therapeutic management, and follow-up. In this review, imaging data for renal
masses that urologists need for accurate treatment planning will be discussed. The role of US, CEUS, CT and
mpMRI in the detection and characterization of renal masses, RCC staging and follow-up of surgically treated or
untreated localized RCC will be presented. The role of percutaneous image-guided ablation in the management of
RCC will be also reviewed.

1. Introduction [12,13].
Despite their lower incidence compared to cystic masses, up to 90 %
The wide availability of cross-sectional imaging is responsible for the of solid renal tumours are malignant, and RCC accounts for 90 % of renal
increased detection of small, usually asymptomatic renal masses [1–7]. malignancies [2,3]. The commonest benign solid renal masses are
Most renal masses are benign cysts. However, renal cell carcinoma angiomyolipoma (AML) and renal oncocytoma [2–7]. AMLs without
(RCC) is often detected incidentally, and patient prognosis is better in visible fat (fat-poor AMLs) account for approximately 5 % of AMLs
these cases [8–11]. Accurate noninvasive renal mass characterization is [2–7].
important to ensure proper treatment planning [1–7]. RCC prognosis is primarily related to tumour stage, histologic sub­
The first step when detecting a renal mass is to differentiate between type, and nuclear grade [2–4]. Preoperative RCC staging and anatomic
benign cysts and solid masses. Most cystic renal masses are benign, and information is vital to guide treatment decisions [14,15]. RCC histologic
when malignant, they are often indolent. Bosniak classification of cystic classification is also important, considering the prognostic and thera­
renal lesions was recently updated [12,13]. Although validation is peutic implications of RCC histologic subtypes. Clear cell RCC, papillary
needed, the primary modifications incorporate cystic masses detected at RCC and chromophobe RCC are the commonest RCC histologic subtypes
MRI or US, establish definitions for previously vague imaging terms and [16]. RCC grading represents another important prognostic factor. The
enable the downgrading of a greater proportion of renal cystic masses four-tiered WHO/ISUP (International Society of Urological Pathology)

Abbreviations: RCC, renal cell carcinoma; CEUS, contrast-enhanced US; mpMRI, multiparametric MRI; AML, angiomyolipoma; ISUP, International Society of
Urological Pathology; TNM, Tumor-Node-Metastasis; CECT, contrast-enhanced CT; NPV, negative predictive value; PPV, positive predictive value; DWI, diffusion-
weighted imaging; VHL, von Hippel-Lindau; MDT, Multidisciplinary Team Meeting.
* Corresponding author at: Department of Clinical Radiology, School of Health Sciences, Faculty of Medicine, University of Ioannina, University Campus, 45110,
Ioannina, Greece.
E-mail addresses: a_tsili@yahoo.gr (A.C. Tsili), andrima@otenet.gr (E. Andriotis), myrgel@gmail.com (M.G. Gkeli), mkrokidis@med.uoa.gr (M. Krokidis),
mystas@otenet.gr (M. Stasinopoulou), medvark3@yahoo.com (I.M. Varkarakis), lmoulop@med.uoa.gr (L.-A. Moulopoulos).

https://doi.org/10.1016/j.ejrad.2021.109777
Received 7 April 2021; Received in revised form 9 May 2021; Accepted 14 May 2021
Available online 15 May 2021
0720-048X/© 2021 Elsevier B.V. All rights reserved.
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

grading system has replaced the traditional Fuhrman grading system Table 1
[17]. RCC TNM classification system [15].
Surgery, including radical nephrectomy or partial nephrectomy is T - Primary Tumor
the recommended treatment for localized RCC [2,18]. Elderly and pa­
TX Primary tumor cannot be assessed
tients with comorbidities and incidental small renal masses have a low T0 No evidence of primary tumor
RCC-specific mortality and significant competing-cause mortality, T1 Tumor ≤ 7 cm in greatest diameter, limited to the kidney
therefore ablative techniques and active surveillance may represent T1a Tumor ≤ 4 cm
alternative treatment options [2,18–20]. Active surveillance defined as T1b Tumor > 4 cm but ≤ 7 cm
T2 Tumor > 7 cm in greatest diameter, limited to the kidney
the initial monitoring of tumor size by serial imaging, with delayed T2a Tumor > 7 cm but ≤ 10 cm
intervention reserved for tumors showing clinical progression during T2b Tumor > 10 cm, limited to the kidney
follow-up, represents a safe management option, not compromising T3 Tumor extends into major veins or perinephric tissues, but not to the ipsilateral
oncologic outcomes [2,18,21,22]. adrenal gland and not beyond Gerota’s fascia
T3a Tumor grossly extends into the renal vein or its segmental (muscle-containing)
Imaging is pivotal in the diagnosis and characterization of renal
branches, or invades perirenal and/or renal sinus fat, but not beyond Gerota’s fascia
masses, providing valuable information regarding staging, prognosis, T3b Tumor grossly extends into inferior vena cava below diaphragm
therapeutic management, and follow-up [7–9,23–25]. US can easily T3c Tumor grossly extends into inferior vena cava above the diaphragm or invades
characterize most incidental renal masses as simple cysts, but it cannot the wall of the vena cava
always differentiate between benign and malignant solid renal tumors T4 Tumor invades beyond Gerota’s fascia (including contiguous extension into the
ipsilateral adrenal gland)
[26]. Contrast-enhanced ultrasonography (CEUS) has emerged as a N - Regional Lymph Nodes
valuable additional tool, useful in differential diagnosis between solid NX Regional lymph nodes cannot be assessed
renal masses and pseudotumors, and between complex renal cysts and N0 No regional lymph node metastasis
solid renal tumors [26–29]. CT represents the gold standard for the N1 Metastasis in regional lymph node(s)
M - Distant Metastasis
characterization of renal masses and for RCC staging [14,30]. Multi­
M0 No distant metastasis
parametric MRI (mpMRI) represents a useful adjuvant tool in the diag­ M1 Distant metastasis
nostic armamentarium of renal masses [31,32].
In this review, we comment on the role of multimodality imaging,
including US, CEUS, CT and mpMRI in the management of renal masses. Imaging plays an important role in providing the above information,
Specifically, the role of imaging modalities in the characterization of predicting the forthcoming difficulties of such surgery.
cystic renal masses, differentiation between RCC histologic phenotypes Various nephrometry scores (RENAL, Padua, c-index) have been
and common benign renal tumors, RCC grading, RCC staging and follow- used to quantify this expected difficulty [34]. The RENAL nephrometry
up of treated or untreated localized RCC is reviewed. The role of mini­ score, assigns tumor with a score depending on points gathered from the
mally invasive, image-guided curative management of RCC is addressed. (R)adius of the tumor, the (E)xophytic/endophytic nature, the (N)
Urologist’s perspective on requirements regarding the radiology report earness to the collecting system or sinus, the (A)nterior(a)/posterior(p)
on renal masses is presented. descriptor, and the (L)ocation relative to the polar line. Various publi­
cations have confirmed correlation of RENAL nephrometry score with
2. What the urologist expects from the radiologist surgical decision making, surgical complications, postoperative func­
tional outcomes, histologic factors such as stage, grade, and
Despite excellent reported sensitivity and specificity for cross- cancer-specific survival rates [34].
sectional imaging in the detection of renal masses, occasionally, small During partial nephrectomy, renal tumor must be identified and
(< 2 cm), usually endophytic renal tumors may be missed [33]. Potential resected. This requires removal of the perinephric fat, which can
implications for the treating urologist may exist, if a radiology report sometimes be tedious. Imaging may predict this difficulty by measuring
misses the diagnosis. perinephric fat thickness, particularly medial and posterior perinephric
The probability of a renal mass being malignant is inversely pro­ fat, and reporting perinephric fat stranding [35]. Of equal importance is
portional to its size. Since more than 50 % of newly diagnosed renal the position of the kidney in relation to the thoracic cage, especially for
masses are less than 3 cm, it is obvious that the urologist would want to open surgery. Information regarding vascular anatomy, and specifically,
be certain of mass histology, before deciding treatment. Unfortunately, the origin, number, division, and course of renal arteries and veins is
histologic characterization based on imaging criteria alone is not always also essential [36].
possible. Understandably, this is a difficult task, since even renal tumor Imaging provides a plethora of information to the urologist necessary
biopsy can be nondiagnostic in up to 8 % of cases, in centers of excel­ for treatment planning, therefore, the radiology report is invaluable and
lence [33]. Fat-poor AMLs, small papillary RCCs and renal oncocytomas should be provided, preferably in a structured format (Table 2) [36].
pose a diagnostic challenge to both radiologists and pathologists.
Imaging is cardinal for RCC staging. Table 1 shows the RCC Tumor- 3. Ultrasonography/contrast-enhanced ultrasonography
Node-Metastasis (TNM) classification system [2,15]. The upper limit of
the inferior vena cava neoplastic thrombus guides surgery, since a Ultrasonography represents the first-line imaging modality for the
thrombus reaching the thorax and heart requires cardiothoracic sur­ investigation of suspected renal disease [26–29]. US may reliably
geons and special anesthetic requirements. Exact regional lymph node differentiate between cystic and solid renal lesions and may characterize
extension mapping will help the surgeon remove palpable lymph nodes. minimally complex renal cysts [28]. US is recommended for the
Removal of the ipsilateral adrenal gland is probable if imaging implies assessment of indeterminate homogeneous, hyperdense renal masses,
invasion by the neighboring tumor. Number, size, and location of distant incidentally found at CT, measuring 20-70HU on unenhanced images or
metastases will predict technical feasibility of metastasectomy and help more than 20HU on single-phase contrast-enhanced images [6]. These
decide whether to proceed or not with cytoreductive nephrectomy [2]. lesions often represent benign hemorrhagic/proteinaceous cysts and can
Although a tumor size of 4 cm is the accepted limit for partial ne­ be safely characterized with US, with a reported sensitivity and speci­
phrectomy, other factors may allow partial resection of a larger tumor or ficity of 81.8 % and 92.9 %, respectively [27]. US also remains the
dictate radical removal of a smaller one. Therefore, information primary imaging modality for the detection and preliminary evaluation
regarding the relationship of the tumor to the collecting system, its vi­ of solid renal tumors [28]. However, the technique cannot always
cinity to the renal hilum and the endophytic or exophytic nature of the accurately differentiate between benign and malignant solid renal tu­
tumor, may define the difficulty of performing partial nephrectomy. mors [26,28,29].

2
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

Table 2
Suggested CT report template for preoperative assessment of solid renal masses,
suspected or proven to represent RCC [36].
Kidney unit with tumor
Nephrometry score
• Tumor radius (cm)
• Exophytic > 50%, Exophytic < 50%, Endophytic
• Distance to the collecting system [> 7, 4-7, < 4] (mm)
• Location: Anterior/Posterior (A/P/X)
• Location regarding to the Polar lines (above/below/cross/ between)
• Extension into renal vein
Extrarenal structures adjacent to the lesion (distance to the nearest anatomic
structure and renal hilar vasculature)
Perinephric fat stranding
Amount of perinephric visceral fat [scant/abundant]
Extension of tumor into perirenal fat, pararenal fascia, adrenal gland
Kidney location [standard, high, low, ectopic]
Kidney size
Collecting system [standard, duplicated]
Vessels
• distance of main renal artery origin to the first branch and to the renal hilum
• accessory renal arteries
• length from inferior vena cava to right renal hilum
• length from aortic edge to the left hilum
• accessory renal veins
Parenchymal variant anatomy [standard, dromendary humb, fetal lobulation,
column of Bertin, renal cleft, congenital fusion/rotation]
Benign pathology [cysts, stones/calcifications, scars, AML]
Contralateral kidney
Kidney size
Enhancement [normal, delayed]
Pathology [cysts, stones/calcifications, scars, AML]
Other
Regional lymphadenopathy [location, size, correlation with large vessels]
Distant metastases

CEUS with its lack of nephrotoxicity, absence of ionizing radiation,


and the ability to evaluate enhancement patterns, can accurately char­
acterize many renal lesions, without the need of CT or MRI [26–29].
Main renal applications include differentiation between solid renal tu­
mors and pseudotumors, characterization of complex cystic renal
masses, characterization of indeterminate renal masses and follow-up of
non-surgically treated renal masses [29].
After ultrasound contrast agent injection, the renal artery and main
branches enhance first, followed rapidly by the segmental, interlobar,
arcuate and interlobular arteries. Subsequently, complete cortical
enhancement is seen (cortical phase, 15− 30 s), followed by medullary
enhancement (parenchymal phase, where both cortex and medulla
enhance homogeneously, 25sec-4 min). As ultrasound contrast agents
are not excreted by kidneys, no excretory phase is obtained [37–41].
CEUS is strongly recommended to differentiate between renal tumors Fig. 1. Sagittal (a) grayscale and (b) power Doppler images depict a solid left
and pseudotumors (prominent columns of Bertin, dromedary or splenic renal lesion (arrows) in the interpolar region, mainly isoechoic, when compared
humps, persistent fetal lobulations and areas of renal parenchyma to normal renal parenchyma. The mass appears to displace the surrounding
adjacent to cortical scarring, which display compensatory hypertrophy), blood vessels. After ultrasound contrast agent injection (c) the lesion (arrows)
not characterized with conventional US, with an accuracy up to 95 % shows similar enhancement as the remaining renal parenchyma, a finding
[26,29]. The criterion to diagnose pseudotumor is the demonstration of suggestive for the presence of renal pseudotumor. (Courtesy: Dr. DD. Kokkinos).
the same enhancing pattern as the surrounding parenchyma in all phases
(Fig. 1) [26,29,41]. On the contrary, enhancement of most solid renal non-surgical complex cystic renal lesions [26,29,53].
tumors differs from the surrounding renal parenchyma, with a difference CEUS has an important role in the characterization of indeterminate
in the degree or distribution of enhancement at least in one renal lesions, including avascular renal masses, without the typical US
contrast-enhanced phase [29,41,42]. Solid renal tumors do not display findings of a simple cyst, complex renal cysts and masses with equivocal
specific perfusion patterns, therefore their accurate characterization by CT enhancement or indeterminate CT findings [6,26–29,54–56]. In a
CEUS is often not possible [23,40,41,43–46]. retrospective study, CEUS had a sensitivity of 100 %, specificity of 95 %,
CEUS can be used to characterize complex cystic renal masses, with positive predictive value (PPV) of 94.7 %, and negative predictive value
accuracy comparable to that of CT and MRI and reported concordance (NPV) of 100 % in the characterization of indeterminate renal masses
rates between the three imaging techniques approximately of 90 % [55]. CEUS follow-up is also recommended in these cases [29].
(Fig. 2) [26,29,47–51]. In addition, CEUS has been reported more sen­ CEUS may highlight renal vein invasion by RCC at the initial eval­
sitive than CT in demonstrating minimal wall and/or septal enhance­ uation, as arterial thrombus vascularization differentiates bland
ment and solid, enhancing components in a complex cystic renal mass thrombus from a neoplastic one, with a reported sensitivity, specificity,
[26,29,51,52]. CEUS is also recommended for the follow-up of PPV, NPV and accuracy of 83 %, 96 %, 71 %, 98 % and 94 %, respec­
tively [14,27,29,57]. Finally, CEUS is helpful in the follow-up of

3
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

Fig. 2. (a) Grayscale US image demonstrates a right renal cystic lesion with an irregularly thickened internal septum (arrow). (b) Contrast-enhanced US shows this
part enhances (arrows); therefore, the lesion is characterized as Bosniak class III. (Courtesy: Dr. DD. Kokkinos).

post-ablated RCC, allowing the detection of intralesional enhancement,


Table 3
indicative of residual or recurrent tumor, with a sensitivity, specificity,
Clinical indications for renal US and contrast-enhanced US [41].
PPV, NPV, and overall accuracy of 96.6 %, 100 %, 100 %, 95.8 %, and
98.1 %, respectively [18,29,58]. Table 3 shows main clinical indications Clinical indications Color Doppler US Contrast-enhanced US
findings findings
for renal US and CEUS.
Nevertheless, the widespread use of CEUS in everyday practice is still Differential diagnosis may be nonspecific renal tumor vascularity
questionable. Continuing training, experience, revising of Bosniak between solid renal differs from that of normal
tumors and parenchyma, at least in one
classification, additional research for including CEUS in clinical uro­ pseudotumors post-contrast phase/
logical guidelines and new technologies, such as, fusion imaging may pseudotumors enhance
improve the diagnostic accuracy of the technique, guarantee its safety, parallel to renal
and confirm its role in the management of renal masses [59–62]. parenchyma in all phases
Characterization of solid often nonspecific Color often nonspecific
renal tumors Doppler has limitations neoplastic renal vein
4. Computed tomography in assessing neoplastic thrombus enhances/bland
invasion of the renal thrombus shows lack of
Contrast-enhanced CT (CECT) represents the gold standard imaging vein in RCC enhancement
Differentiation between limitations in assessing solid hypovascular renal
technique for the detection and characterization of renal masses [1–7,
cystic and solid renal possible perfusion in tumors enhance, even
23–25,30,63–65]. Renal CT protocol consists of an unenhanced phase, masses echogenic cysts slightly/debris do not
combined with one or more contrast-enhanced phases, namely the enhance superior to CT/
corticomedullary phase (40− 70 sec), the nephrographic phase MRI in diagnosing cystic
(100− 120 sec) and the delayed-excretory phase (7− 10 min) [66]. The RCC
Characterization of limitations in assessing depicts wall/septal and/or
nephrographic phase is optimal for RCC detection, as tumor contrast complex cystic renal possible perfusion in nodules enhancement
washout becomes visible, against the homogeneously enhancing renal masses septa and/or cyst similar or higher accuracy
parenchyma [30,67,68]. The corticomedullary phase may help in nodules compared to CT for renal
sub-typing RCC, as clear cell RCC enhances avidly in this phase [66]. cystic lesions classification,
according to Bosniak
CECT is the modality of choice for the evaluation of cystic renal
criteria
masses [12,13,69–72]. Table 4 shows CT characteristics of renal cystic RCC post-ablation no role confirms treatment results
masses based on the recent Bosniak classification [12,13]. A homoge­ similar accuracy to that of
neous renal mass measuring between -10HU and +20HU on unenhanced CT/MRI post-ablation
CT corresponds to a simple benign cyst, without the need for additional contrast-enhancing areas
are considered residual or
imaging. A homogeneous hyperdense lesion measuring more than 70HU recurrent tumor
on non-contrast CT, indicates a benign hemorrhagic or proteinaceous
cyst [6]. Lesions either homogeneous or inhomogeneous within the

4
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

Table 4
Bosniak Classification of Cystic Renal Masses, Version 2019 [12,13].
Category CT findings MRI findings

I well-defined cystic mass, well-defined cystic mass,


homogeneous, watery density homogeneous, hyperintense T2
(-10 to +20 HU), smooth, thin signal, similar to cerebrospinal
wall (≤ 2 mm) that may enhance, fluid, smooth, thin wall (≤ 2 mm)
absence of septa and/or that may enhance, absence of septa
calcifications and/or calcifications
well-defined mass, smooth, thin well-defined mass, smooth, thin
wall (≤ 2 mm) wall (≤ 2 mm)
• cystic mass, few (1-3) thin (≤
2 mm) septa; wall and septa
• cystic mass, few (1-3) thin (≤
may enhance; calcifications of
2 mm) contrast-enhancing septa
any type
or any nonenhancing septa; may
• homogeneous hyperdense (≥
contain calcifications of any
70HU) mass at unenhanced CT
type
• homogeneous nonenhancing
II • homogeneous, markedly
mass > 20HU at renal mass
hyperintense T2 signal, similar
protocol CT; may have
to cerebrospinal fluid
calcifications
• homogeneous, markedly
• homogeneous mass -10 to
hyperintense fat-suppressed T1
+20HU at unenhanced CT
signal, equal or 2.5 times higher
• homogeneous mass 21-30HU
than that of normal renal
at portal phase CT
parenchyma
• homogeneous hypodense
mass, too small to characterize
• cystic mass, smooth, minimally
thickened (3 mm) enhancing
cystic mass, smooth, minimally
wall and/or septa, or many (≥
thickened (3 mm) enhancing
IIF 4) smooth, thin enhancing septa
wall and/or septa, or many (≥ 4)
• cystic mass, heterogeneously
smooth, thin enhancing septa
hyperintense fat-suppressed T1
signal
cystic mass, thickened (≥ 4 mm) cystic mass, thickened (≥ 4 mm)
enhancing wall and/or septa, or enhancing wall and/or septa, or
enhancing nodule(s): convex enhancing nodule(s): convex
III protrusion(s) arising from the protrusion(s) arising from the wall
wall and/or septa, irregularly and/or septa, irregularly
thickened (≤ 3 mm), with obtuse thickened (≤ 3 mm), with obtuse
margins margins
cystic mass, one or more cystic mass, one or more
enhancing nodule(s): convex enhancing nodule(s): convex
protrusion(s), arising from the protrusion(s), arising from the wall
IV
wall and/or septa, ≥ 4 mm, with and/or septa, ≥ 4 mm, with obtuse
obtuse margins, or of any size, margins, or of any size, with acute
with acute margins margins

20-70HU range, are considered indeterminate and require further


evaluation [6]. The most commonly used method to characterize inde­
terminate renal masses, including both cystic and solid renal lesions is
CECT [5,6]. Enhancing solid renal tumours or enhancing components in
cystic renal masses (Fig. 3) are strongly suggestive of malignancy [65].
Differences of at least 10HU between unenhanced and CECT have been
proposed to differentiate renal cysts from solid masses [6]. However,
more conservative values, such as 15-20HU are often used to account for
partial volume-averaging artifacts [73].
Because CT is considered the gold standard for the detection of renal
masses, the accuracy of the technique in the detection of renal tumors is
difficult to define, based on literature data [30,74,75]. In a retrospective
study of small (≤ 4 cm) indeterminate renal masses, CECT accuracy in
predicting RCC diagnosis was 79.4 % [74]. Moreover, CT evaluation of
small (≤ 1.5 cm) renal masses may be problematic, due to lesion pseu­
doenhancement and partial volume-averaging artifacts, which limit the
assessment of contrast enhancement [76–78].
CT findings related to lesion homogeneity and enhancement patterns
Fig. 3. Clear cell RCC of the left kidney. (a) Coronal and (b) sagittal contrast-
are reported to correlate with RCC histologic subtypes [30,79–81]. enhanced CT images during the corticomedullary phase depict a <50 % exo­
Typical clear cell RCC enhances strongly in the corticomedullary phase phytic left renal cystic lesion (arrow), with a 5 mm contrast-enhancing nodule
(114 ± 44 HU), with a rapid washout in the nephrographic phase (long arrow, b, Bosniak class IV). Left simple benign renal cyst (asterisk). (c)
(66 ± 24 HU) [30,79]. Large clear cell RCCs are often heterogeneous, Coronal three-dimensional maximum intensity projection reconstructed image
due to the presence of necrosis and/or cystic degeneration [79]. Papil­ shows distance of the left renal artery origin to the first branch (arrow).
lary RCC is usually small, homogenous and hypovascular, with weak

5
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

enhancement in the corticomedullary phase, up to 20 HU [30,68].


Absence of enhancement is seen in up to 25 % of papillary RCCs, and
further evaluation with CEUS or mpMRI is advised to verify the presence
of an enhancing solid tumor [30].
Chromophobe RCC and renal oncocytoma share similar histologic
and imaging features [31,32,82–84]. Therefore, differential diagnosis is
often difficult and histologic exploration may be needed (Fig. 4) [7,31,
32]. Peripheral location, presence of a central stellate scar or
spoke-wheel enhancement and segmental enhancement inversion are
common findings for both entities [7,31,32,82–88]. Most fat-poor AMLs
are hyperdense on unenhanced CT, homogeneously enhancing after
contrast administration [30,63]. However, differentiation between
fat-poor AML and RCC by CT alone is often difficult and further evalu­
ation with mpMRI, biopsy, or even surgical resection may be required
[30,63,89–91].
Recently, contrast-enhanced dual energy CT has been reported to
improve the diagnostic performance of CT in the characterization of
renal masses, with sensitivity and specificity greater than 95 % in the
evaluation of renal tumors [92,93]. New techniques, including CT Fig. 5. Renal incidentaloma detected on US. (a) Unenhanced CT depicts an
perfusion, CT texture analysis and CT-based radiomics may provide isoattenuating renal mass (arrow) in the middle of the right kidney. (b) An
important additional information in renal mass characterization inhomogeneous enhancement pattern (arrow) is seen in the corticomedullary
[94–99] phase. The lesion (arrow) has a subtle washout effect (c) in the nephrogenic
phase, becoming more intense (d) in the excretory phase. Renal tumor biopsy
Up to now, multimodality imaging cannot always accurately char­
showed renal oncocytoma.
acterize renal masses discovered incidentally, especially, small renal
masses (≤ 4 cm) [100]. Percutaneous renal tumor biopsy has been
shown to help avoid surgery in up to 33 % of cases initially considered to separate procedure or just prior to treatments [102]. Biopsies can be
be malignant, based on imaging [101]. Indications for renal tumor bi­ performed under US or CT-guidance.
opsy in localized RCC include small renal masses, when the results may CECT is widely accepted as the diagnostic modality of choice for RCC
alter management (Fig. 5) and masses with findings suggestive of lym­ staging, with sensitivity and specificity up to 90 % [2–4,14,103,104].
phoma, metastasis, infectious or inflammatory lesions [102]. Renal RCC size and degree of local invasion determine the T stage (Table 1).
tumor biopsy is also recommended before thermal ablation, as a Although, some studies have reported that CT often overestimates tumor

Fig. 4. Clear cell RCC in a woman with a history of breast cancer. (a) Axial unenhanced CT shows a left renal mass (arrow). The lesion has a CT density mainly
similar to that of normal renal parenchyma and small calcifications. (b) In the corticomedullary phase, the lesion (arrow) shows strong, heterogeneous enhancement,
with a washout effect in the nephrographic phase (c). (d) Renal tumor biopsy revealed a clear cell RCC.

6
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

size, compared to surgical specimen, discrepancies are minimal and


clinically insignificant in most cases [105,106]. Perinephric or renal
sinus fat invasion is not always easily detected on CT [107]. Thin sec­
tions and multiplanar reformations have improved the detection of
perinephric fat infiltration, although false-positives, due to inflamma­
tion, oedema, vascular engorgement, or fibrosis may be encountered
[107–109]. CT is highly accurate in the diagnosis of RCC spread into the
renal vein, with a NPV of 97 % and PPV of 92 % [14,110]. CT is also
effective in assessing the superior extent of inferior vena cava thrombus
[111].
CT offers a high NPV in excluding adjacent organ involvement Fig. 6. Follow-up unenhanced MRI in a patient with renal failure and a history
beyond the Gerota΄s fascia or involvement of the ipsilateral adrenal of right radical nephrectomy, due to RCC. T2-weighted imaging in (a) axial and
(b) coronal planes show neoplastic thrombus (arrow) into the inferior
gland [14,112,113]. In a retrospective observational study, CT had a
vena cava.
sensitivity of 100 %, specificity of 95.2 % and NPV of 100 % in diag­
nosing adrenal gland invasion [113]. However, CT cannot always
discriminate abutment from direct invasion [14]. due to its advantages over CT, namely decreased sensitivity to calcifi­
CT tends to overdiagnose lymph node spread, which is defined as a cation, increased sensitivity to enhancement and absence of pseu­
lymph node with a short axis diameter greater than 1 cm and disruption doenhancement, may provide additional diagnostic information in the
of normal lymph node architecture. However, based on size criterion, CT assessment of renal cystic masses, with abundant thick or nodular cal­
has 10 % of false-negatives, while approximately 50 % of enlarged cifications at CT, hyperdense, homogeneous non-enhancing renal tu­
lymph nodes are proved benign [14,114]. Distant metastases from RCC mors, larger than 3 cm and heterogeneous, non-enhancing masses at CT
are often found in the lungs, bones, liver, and brain and less commonly [12,13].
in the thyroid, pancreas, muscle, skin, and soft tissue [14,115]. Chest CT CT, MRI and CEUS represent equivalent alternatives for the initial
is useful to detect lung metastases and metastatic mediastinal lymph assessment of an indeterminate renal mass [6]. MRI is primarily rec­
nodes and is mainly recommended in large-sized RCCs [14]. Visceral ommended for the assessment of solid renal masses with inconclusive CT
metastases tend to be hypervascular and corticomedullary phase may findings, especially those smaller than 1.5 cm [5,6,125,126].
help detect arterial enhancement [14,115]. Multiparametric MRI features, including T2 signal, chemical shift
Multiplanar reformations and three-dimensional reconstructions imaging characteristics, apparent diffusion coefficient (ADC) signal,
greatly help in preoperative planning, providing important anatomic patterns of early enhancement and de-enhancement help in the char­
information, such as kidney position relative to the surrounding bones, acterization of solid renal tumors (Table 5) [7,31,32,63,127–129]. Clear
tumor location and depth of extension into the kidney, relationship of cell RCC is often large and heterogeneous, with high T2 and ADC signal,
the tumor to the renal collecting system, and accurate delineation of the and early, strong heterogeneous enhancement, followed by rapid
arterial and venous anatomy [116,117] Preoperative CT also provides de-enhancement (Fig. 7) [7,31,32,63,127–132]. Tumor pseudocapsule
information on the morphology and function of the contralateral kidney. is often detected as a low T2 signal halo around the neoplasm (Fig. 7b)
Synchronous primary tumors should be searched for, as RCC can be and suggests kidney-confined disease [133]. Multiparametric MRI has
multifocal [14]. sensitivity and specificity up to 92 % and 83 %, respectively in the
CT is indicated for RCC follow-up after surgery or post-ablation, in diagnosis of clear cell RCC [32].
moderate to high-risk patients [18]. A baseline chest and abdominal CT Papillary RCC is often small, peripherally located, with low T2 and
within three to six months after surgery is recommended, followed by ADC signal and slow, progressive enhancement. Intratumoral hemor­
imaging every six months for at least three years and annually there­ rhage is often detected, either as a hyperintense T1 area or as a signal
after, for two years [118]. drop on in-phase imaging, corresponding to prolonged or chronic
hemorrhage [31,32,68,134–137]. A recently published retrospective
study of 109 resected small renal tumors reported a diagnostic accuracy,
5. Multiparametric MRI
of 81 % and 91 % in the characterization of clear cell RCC and papillary
RCC, respectively using mpMRI, with a moderate to substantial
Multiparametric MRI represents a valuable complementary imaging
inter-reader agreement among seven radiologists [135]. Chromophobe
technique for the assessment of renal masses, providing both morpho­
RCC usually has intermediate to low T2 signal, a low ADC when
logic and functional information [5–7,12–14,18,31,32]. MRI is mainly
compared to clear cell RCC, and, intermediate early enhancement, fol­
recommended when optimal CT cannot be performed (severe allergy to
lowed by gradual de-enhancement [31,32,82,134].
iodinated CT contrast or high risk for contrast-induced nephropathy) or
Multiparametric MRI often enables the characterization of fat-poor
when radiation exposure is contraindicated (young age or pregnant
AML, with sensitivity and specificity up to 100 % and 89 %, respec­
women) [5–7,12–14,18,31,32]. However, gadolinium-based MR agents
tively [31]. Fat-poor AML is characteristically small, homogeneously
should be administered with caution during pregnancy, only when there
hypointense on T2-weighted imaging and ADC map, with avid, early
is a very strong clinical indication and the potential benefits justify the
enhancement and rapid washout. Intratumoral fat is rarely detected, as a
potential unknown risk to the fetus [119]. When both iodinated CT
signal drop on opposed-phase imaging [31,134,138]. Common mpMRI
contrast and gadolinium-based MR agents cannot be used, MRI without
findings of renal oncocytoma include the following: peripheral location,
iv contrast is highly recommended for renal mass characterization, RCC
intermediately high T2 signal, high ADC, strong, early enhancement and
staging and follow-up (Fig. 6) [5,6,12–14,18].
gradual de-enhancement [31,32,139].
Renal MRI protocol includes the following sequences: T1-weighted
Results on the efficacy of DWI in the characterization of solid renal
with in-phase and opposed-phase imaging (or DIXON technique), T2-
masses are conflicting, with significant ADC overlapping among
weighted, diffusion-weighted imaging (DWI), subtracted dynamic
different histologic subtypes. However, DWI may be used to predict RCC
contrast-enhanced and delayed, post-contrast T1-weighted imaging
histologic grade, with restricted diffusion often seen in high-grade clear
[120].
cell RCC, when compared to low-grade tumors [140–145].
Although CT and MRI have similar accuracy in the characterization
A variety of new MRI techniques, including intravoxel incoherent
of cystic renal masses, mpMRI may show additional findings, such as
motion, diffusion kurtosis, texture analysis, arterial spin labelling, blood
more septa, wall and/or septal thickening and contrast enhancement,
oxygenation level-dependent MRI and radiomics have been introduced
which may result in lesion upgrading (Table 4) [5,6,12,121–124]. MRI,

7
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

Table 5 6. Minimally invasive, image-guided, curative management of


Multiparametric MRI findings useful to differentiate common solid renal tumors RCC
(mpMRI: multiparametric MRI; RCC: renal cell carcinoma; ADC: apparent
diffusion coefficient) [31,32]. Advanced-stage RCC has a clear management pathway that mainly
Histologic type mpMRI features consists of radical nephrectomy or palliative treatment. The manage­
clear cell RCC • large size ment of small, early-stage, asymptomatic, sporadic RCC is a bit more
• heterogeneous complex, given that the behavior of the lesion cannot be predicted. The
• necrosis: hyperintense T2 area, absence of general consensus is to assess growth pattern with active surveillance.
enhancement When treatment is considered, open or laparoscopic partial nephrec­
• high T2 signal
• pseudocapsule
tomy appears to be the standard approach, aiming to preserve as much
• may signal drop on opposed phase healthy parenchyma as possible.
• high ADC Interventional Oncology may offer another minimally invasive
• high perfusion locoregional treatment for such patients with percutaneous image-
• rapid washout
guided ablation [19,20,155]. Percutaneous ablation is an established
papillary RCC • small size < 3 cm
• sharply defined form of treatment used in a number of other organs [156]. The tech­
• homogeneous nology that was initially implemented for the ablation of renal tumors
• peripheral location was radiofrequency [157]. The approach was triggered by the necessity
• low T2 signal to treat patients that were poor surgical candidates or those who have a
• pseudocapsule
limited life expectancy for other reasons with an effective minimally
• hemorrhage: hyperintense T1 or fat-suppressed T1 area
or signal drop on in phase invasive procedure. The thermal effect of radiofrequency is based on
• rarely, signal drop on opposed phase high-frequency electric current that causes oscillation of the tissue
• low ADC molecules and produces heat [158]. When temperature exceeds 60 ◦ C,
• low perfusion
the tissue is destroyed. This technology is now established in the treat­
chromophobe RCC • large size (mean diameter, 7.2 cm)
• sharply defined ment of small renal masses, with excellent long-term results [159,160].
• homogeneous, absence of necrosis Cryoablation is another modality that is established for the percuta­
• peripheral location neous image-guided treatment of RCC. The physical principle of cry­
• central scar, spoke-wheel enhancement, segmental oablation is based on the Joule-Thompson principle that describes the
enhancement inversion
temperature drop after the rapid expansion of Argon. Cryoablation le­
• intermediate T2 signal
• low ADC thal effect is based on direct cellular damage caused from osmotic
• intermediate perfusion cellular dehydration due to extracellular freezing and due to the intra­
fat-poor • small size < 4 cm cellular formation of ice [161]. Percutaneous cryoablation has also
angiomyolipoma • angular interface, ‘ice-cream cone’ sign
offered excellent long-term results in the management of small renal
• low T2 signal
• rarely, signal drop on opposed phase
masses [162,163]. Furthermore, microwave ablation has recently been
• low ADC introduced in the percutaneous RCC treatment and has already pro­
• high perfusion duced some encouraging results [164]. Microwave is based on the use of
• rapid washout an electromagnetic wave that causes continuous rotation of tissue water
renal oncocytoma • peripheral location
molecules. The non-equal distribution of electric charge of the water
• sharply defined
• central scar, spoke-wheel enhancement, segmental molecules causes their continuous re-orientation within the oscillating
enhancement inversion field; this movement increases their kinetic energy and is deposited in
• intermediate T2 signal the tissue as thermal energy [164].
• may signal drop on opposed imaging
Minimally invasive percutaneous ablation has a significant role in
• high ADC
• high perfusion
patients with multiple, bilateral synchronous RCCs, such as in von-
Hippel-Lindau (VHL) syndrome [165]. Another group of patients that
may benefit significantly from percutaneous ablation are patients that
for renal mass characterization, evaluation of RCC aggressiveness, and underwent nephrectomy and develop a new RCC or a metastatic lesion
assessment of treatment response, although validation is needed in the contralateral kidney or those who develop a RCC in a single
[146–151]. Moreover, investigation of the biochemical environment of functioning kidney [166]. For such patients, percutaneous ablation of­
renal masses by proton MR spectroscopy may additionally help in lesion fers a valid tumor control solution, with preservation of renal function
characterization [152]. [166–168].
CT and MRI represent equivalent alternatives for RCC staging, with Even though there are at least three ablation modalities used for the
similar diagnostic performances [14]. MRI may provide additional in­ percutaneous locoregional treatment of RCC there is no evidence of
formation in cases with indeterminate CT findings, especially when superiority of one of the three until today regarding complications,
assessing perinephric fat infiltration, extent of neoplastic thrombus into postprocedural renal function tests, local tumor progression, or cancer-
the renal vein and/or the inferior vena cava, and possible invasion of the specific survival [169–173].
inferior vena cava wall by RCC thrombus [14,153,154]. One of the main advantages of percutaneous ablation is that the level
Both CT and MRI perform well in the follow-up of RCC after surgery of sedation may be individualized and it may be safely performed under
or post-ablation, with a high accuracy in the detection of local recur­ moderate sedation, monitored anesthesia, or if required, general anes­
rence and/or distant metastases [18]. In low-risk RCC, MRI may be used thesia. [20,174].
as the primary modality for follow-up, due to the absence of radiation The decision to proceed needs to be obtained via a multidisciplinary
exposure [2]. When intravenous contrast is contraindicated, MRI meeting. The performing interventional radiologist needs to consult the
follow-up without iv contrast is recommended (Fig. 5) [18]. CT, MRI and patient prior to the procedure, in order to discuss the risks and the
CEUS are usually recommended for active surveillance of localized RCC benefits of the procedure, the anesthesia options and to assess the fitness
[18]. of the patient. Anticoagulation needs to be stopped, as per standard
percutaneous procedures and in case of the use of Warfarin, bridging
with heparin is required [175].
Imaging guidance may be performed with a variety of modalities

8
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

Fig. 7. (a) Coronal T2-weighted image depicts a heterogeneous right lower pole renal mass (arrow), of high T2 signal. A hyperintense intratumoral central area is
seen, due to necrosis. The tumour is surrounded by a low T2 signal halo, corresponding to pseudocapsule, on pathology. Axial (b) in-phase and (c) opposed-phase T1-
weighted images shows signal drop (arrow) on the opposed imaging, due to the presence of intratumoral fat. (d) Axial ADC map (b = 600 s/mm2). The mass (arrow)
has a signal similar to that of the surrounding renal parenchyma. (e) Coronal subtracted dynamic contrast-enhanced image shows tumor (arrow) enhancing strongly
and heterogeneously.

including US, CT, MRI, PET, CT-fluoroscopy, or cone-beam CT. De­ prior to MDT discussion and the lesion is biopsied prior to ablation on
cisions regarding imaging guidance should consider patient factors, the same session. In such cases, biopsy should be performed via a coaxial
availability, cost, radiation dose, and operator preference. Navigation system that would also be used as access of the electrode, since bleeding
systems have also been developed, that offer a more accurate needle post biopsy might limit the delineation of the lesion borders [177].
placement and may also predict the ablation area [176] (Fig. 8). The In case of contiguity with the bowel, hydro dissection with non-ionic
patient will be usually admitted in hospital the day of the procedure, will solution needs to be considered, via a thin needle, to displace the bowel
be transferred to a ward post procedure for overnight observation and be and to insulate the lesion [178].
discharged early the next morning, aiming for a less than 24 h stay in Follow-up with triple-phase CT is required four weeks post ablation,
hospital. When the patients are in the CT room, premedication with to assess if there is any residual enhancing tissue. In case of incomplete
1000 mg of Paracetamol intravenously is administered and then, lesion ablation, a second session is required as soon as possible. If the
conscious sedation is given with 1− 4 mg of Midazolam and 50-200mi­ ablation result is satisfactory, with lack of lesion enhancement, then
crograms of Fentanyl, the moment prior to the ablation [20,174]. follow-up with triple-phase CT at six and 12 months is required, and
Biopsy of renal lesions needs to be obtained, prior to ablation in all yearly after, for a total of five years [179–181].
cases. Usually this is obtained prior to any discussion in the Multidis­ Ablation offers excellent long-term oncologic results in the locore­
ciplinary Team Meeting (MDT) for sporadic lesions. For patients with gional treatment of RCC and needs to be offered to every patient with
VHL or patients with known contralateral tumor, no biopsy is required tumor up to 4 cm in diameter. More specifically, the reported primary

9
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

Fig. 8. CT-guided percutaneous ablation of a small endophytic left


renal tumor in a male previously diagnosed with clear cell RCC of
the contralateral kidney, that was treated with radical nephrec­
tomy. Five years later, he presented a new primary 1.7 cm left
renal mass. Renal tumor biopsy confirmed clear cell RCC of the left
kidney. After Multidisciplinary Team Meeting, decision to proceed
with ablation of the lesion was made. (a) Axial contrast-enhanced
CT scan in the corticomedullary phase that confirms the position of
a small left renal endophytic lesion (arrow). (b) CT-guided navi­
gation system (CAS- OneÒIR, CASCINATION AG, Bern,
Switzerland) is used to target the tumor and predict the ablation
area (green circle). Navigation is very helpful in such case, as the
lesion is challenging to target, due to the small size and the
endophytic growth. (c) An RFA electrode is inserted, and ablation
is performed for 12 min. (d) Follow-up contrast-enhanced CT in the
corticomedullary phase six months later confirms satisfactory
ablation of the area, with lack of enhancement and presence of a
“halo” sign (arrow). Satisfactory result was also confirmed in the
five-year follow-up, with preservation of the renal function.

efficacy for the treatment of T1a tumors ranges between 94.4–98.2 %, Declaration of Competing Interest
with secondary efficacy between 98.5 %–99.1 % [159,162,182].
None.
7. Conclusions
References
Renal masses are nowadays increasingly detected in asymptomatic
individuals, as incidental findings at cross-sectional imaging. Multi­ [1] M. Jayson, H. Sanders, Increased incidence of serendipitously discovered renal
cell carcinoma, Urology 51 (1998) 203–205, https://doi.org/10.1016/s0090-
modality imaging, including US, CEUS, CECT and mpMRI play an 4295(97)00506-2.
important role in the detection and characterization of renal masses, [2] B. Ljungberg, L. Albiges, Y. Abu-Ghanem, K. Bensalah, S. Dabestani,
RCC staging and follow-up of surgically treated or untreated RCC. S. Fernández-Pello, R.H. Giles, F. Hofmann, M. Hora, M.A. Kuczyk, T. Kuusk, T.
B. Lam, L. Marconi, A.S. Merseburger, T. Powles, M. Staehler, R. Tahbaz,
Conventional US often represents the first-line imaging tool for the A. Volpe, A. Bex, European association of urology guidelines on renal cell
assessment of renal pathology. The technique may accurately charac­ carcinoma: the 2019 update, Eur. Urol. 75 (2019) 799–810, https://doi.org/
terize simple renal cysts and minimally complex cysts. CEUS has 10.1016/j.eururo.2019.02.011.
[3] S. Campbell, R.G. Uzzo, M.E. Allaf, E.M. Remer, Renal Mass and Localized Renal
emerged as a powerful additional tool for the characterization of renal Cancer: AUA Guideline, J. Urol. 198 (2017) 520–529, https://doi.org/10.1016/j.
masses. Established CEUS applications include the differentiation be­ juro.2017.04.100.
tween solid renal tumors and pseudotumors, characterization of com­ [4] R.D. Ward, H. Tanaka, S.C. Campbell, E.M. Remer, 2017 AUA renal mass and
localized renal Cancer guidelines: imaging implications, Radiographics 38 (2018)
plex cysts and indeterminate renal lesions and follow-up of non-
2021–2033, https://doi.org/10.1148/rg.2018180127.
surgically treated renal masses. CT represents the examination of [5] B.R. Herts, S.G. Silverman, N.M. Hindman, R.G. Uzzo, R.P. Hartman, G.M. Israel,
choice for the assessment of indeterminate renal masses, RCC staging D.A. Baumgarten, L.L. Berland, P.V. Pandharipande, Management of the
and follow-up, in moderate- and high-risk patients. Multiparametric Incidental Renal Mass on CT: A White Paper of the ACR Incidental Findings
Committee, J. Am. Coll. Radiol. 15 (2018) 264–273, https://doi.org/10.1016/j.
MRI is a useful adjunct tool in the management of renal masses, mainly jacr.2017.04.028.
indicated when optimal CT cannot be performed (history of allergy or [6] Expert Panel on Urologic Imaging, Z.J. Wang, P. Nikolaidis, G. Khatri, V.S. Dogra,
renal insufficiency) or when ionizing radiation is contraindicated D. Ganeshan, S. Goldfarb, J.L. Gore, R.T. Gupta, R.P. Hartman, M.E. Heilbrun,
A. Lyshchik, A.S. Purysko, S.J. Savage, A.D. Smith, D.J. Wolfman, J.J. Wong-You-
(young age or pregnancy). The technique may provide additional useful Cheong, M.E. Lockhart, ACR appropriateness criteria® indeterminate renal mass,
information in the characterization of cystic and solid renal tumors, J. Am. Coll. Radiol. 17 (2020) S415–S428, https://doi.org/10.1016/j.
especially of small renal masses. MRI is also recommended for the jacr.2020.09.010.
[7] F.U. Kay, I. Pedrosa, Imaging of solid renal masses, Urol. Clin. North Am. 45
follow-up of low-risk RCC. (2018) 311–330, https://doi.org/10.1016/j.ucl.2018.03.013.
Percutaneous image-guided ablation represents an effective therapy [8] E. Guðmundsson, H. Hellborg, S. Lundstam, S. Erikson, B. Ljungberg, Metastatic
for localized RCC, with acceptable outcome results. Main indications potential in renal cell carcinomas ≤7cm: swedish kidney Cancer quality register
data, Eur. Urol. 60 (2011) 975–982, https://doi.org/10.1016/j.
include the management of small renal masses (≤ 4 cm), multiple RCCs eururo.2011.06.029.
(such as, in VHL syndrome) and RCC in a solitary kidney. [9] A. Volpe, T. Panzarella, R.A. Rendon, M.A. Haider, F.I. Kondylis, M.A.S. Jewett,
A structured template, including the nephrometry score and addi­ The natural history of incidentally detected small renal masses, Cancer 100
(2004) 738–745, https://doi.org/10.1002/cncr.20025.
tional pertinent CT or MRI findings for the reporting of a solid renal
[10] K.H. Tsui, O. Shvarts, R.B. Smith, R. Figlin, J.B. de Kernion, A. Belldegrun, Renal
mass, greatly helps clinicians in precise treatment planning. cell carcinoma: prognostic significance of incidentally detected tumors, J. Urol.
163 (2000) 426–430, https://doi.org/10.1016/s0022-5347(05)67892-5.
Funding [11] J.J. Patard, A. Rodriguez, N. Rioux-Leclercq, F. Guille, B. Lobel, Prognostic
significance of the mode of detection in renal tumours, BJU Int. 90 (2002)
358–363, https://doi.org/10.1046/j.1464-410x.2002.02910.x.
This research did not receive any specific grant from funding [12] S.G. Silverman, I. Pedrosa, J.H. Ellis, N.M. Hindman, N. Schieda, A.D. Smith, E.
agencies in the public, commercial, or not-for-profit sectors. M. Remer, A.B. Shinagare, N.E. Curci, S.S. Raman, S.A. Wells, S.D. Kaffenberger,
Z.J. Wang, H. Chandarana, M.S. Davenport, Bosniak classification of cystic renal
masses, version 2019: an update proposal and needs assessment, Radiology 292
(2019) 475–488, https://doi.org/10.1148/radiol.2019182646.

10
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

[13] S. Krishna, N. Schieda, I. Pedrosa, N. Hindman, R.H. Baroni, S.G. Silverman, M. carcinoma: a systematic review and meta-analysis, Cancer Med. 9 (2020)
S. Davenport, Update on MRI of cystic renal masses including bosniak version 3944–3953, https://doi.org/10.1002/cam4.2993.
2019, J. Magn. Reson. Imaging 2 (2020), https://doi.org/10.1002/jmri.27364. [35] S.M. Lee, I. Robertson, T. Stonier, N. Simson, T. Amer, O.M. Aboumarzouk,
[14] R. Vikram, M.D. Beland, M.D. Blaufox, C.C. Moreno, J.L. Gore, H.J. Harvin, M. Contemporary outcomes and prediction of adherent perinephric fat at partial
E. Heilbrun, S.L. Liauw, P.L. Nguyen, P. Nikolaidis, G.M. Preminger, A.S. Purysko, nephrectomy: a systematic review, Scand. J. Urol. 51 (2017) 429–434, https://
S.S. Raman, M.T. Taffel, Z.J. Wang, R.M. Weinfeld, E.M. Remer, M.E. Lockhart, doi.org/10.1080/21681805.2017.1357656.
ACR appropriateness criteria renal cell carcinoma staging, J. Am. Coll. Radiol. 13 [36] N. Alsaikhan, W. Alshehri, F. Cassidy, L. Aganovic, N. Vahdat, Renal tumor
(2016) 518–525, https://doi.org/10.1016/j.jacr.2016.01.021. structured reporting including nephrometry score and beyond: what the urologist
[15] American Joint Committee on Cancer Kidney, M.B. Amin, S. Edge, F. Greene, et and interventional radiologist need to know, Abdom. Radiol. (NY) 44 (2019)
al. (Eds.), AJCC Cancer Staging Manual, 8th ed, Springer-Verlag, New York, NY, 190–200, https://doi.org/10.1007/s00261-018-1691-0.
2017, pp. 747–756. [37] A. Nilsson, Contrast-enhanced ultrasound of the kidneys, Eur. Radiol. 14 (2004)
[16] H. Moch, A.L. Cubilla, P.A. Humphrey, V.E. Reuter, T.M. Ulbright, The 2016 P104–109.
WHO classification of tumours of the urinary system and male genital organs-part [38] J.M. Correas, M. Claudon, F. Tranquart, O. Hélénon, The kidney: imaging with
a: renal, penile, and testicular tumours, Eur. Urol. 70 (2016) 93–105, https://doi. microbubble contrast agents, Ultrasound Q. 22 (2006) 53–66.
org/10.1016/j.eururo.2016.02.029. [39] C.J. Harvey, A. Alsafi, S. Kuzmich, A. Ngo, I. Papadopoulou, A. Lakhani,
[17] B. Delahunt, J.C. Cheville, G. Martignoni, P.A. Humphrey, C. Magi-Galluzzi, Y. Berkowitz, S. Moser, P.S. Sidhu, D.O. Cosgrove, Role of US contrast agents in
J. McKenney, L. Egevad, F. Algaba, H. Moch, D.J. Grignon, R. Montironi, J. the assessment of indeterminate solid and cystic lesions in native and transplant
R. Srigley, Members of the ISUP Renal Tumor Panel, the International Society of kidneys, Radiographics 35 (2015) 1419–1430, https://doi.org/10.1148/
Urological Pathology (ISUP) grading system for renal cell carcinoma and other rg.2015140222.
prognostic parameters, Am. J. Surg. Pathol. 37 (2013) 1490–1504, https://doi. [40] A. Ignee, B. Straub, G. Schuessler, C.F. Dietrich, Contrast enhanced ultrasound of
org/10.1097/PAS.0b013e318299f0fb. renal masses, World J. Radiol. 28 (2010) 15–31, https://doi.org/10.3233/CH-
[18] Expert Panel on Urological Imaging, A.S. Purysko, P. Nikolaidis, V.S. Dogra, 2010-1352.
D. Ganeshan, J.L. Gore, R.T. Gupta, M.E. Heilbrun, G. Khatri, A.U. Kishan, [41] D.D. Cokkinos, E.G. Antypa, M. Skilakaki, D. Kriketou, E. Tavernaraki, P.
A. Lyshchik, S.J. Savage, A.D. Smith, Z.J. Wang, D.J. Wolfman, J.J. Wong-You- N. Piperopoulos, Contrast Enhanced Ultrasound of the Kidneys: What Is It
Cheong, D.C. Yoo, M.E. Lockhart, ACR appropriateness criteria post-treatment Capable of? Biomed. Res. Int. (2013) 595873, https://doi.org/10.1155/2013/
follow-up and active surveillance of clinically localized renal cell Cancer, J. Am. 595873.
Coll. Radiol. 16 (2019) S399–S416, https://doi.org/10.1016/j.jacr.2019.05.022. [42] S. Mazziotti, F. Zimbaro, A. Pandolfo, S. Racchiusa, N. Settineri, G. Ascenti,
[19] K. Katsanos, L. Mailli, M. Krokidis, A. McGrath, T. Sabharwal, A. Adam, Usefulness of contrast-enhanced ultrasonography in the diagnosis of renal
Systematic review and meta-analysis of thermal ablation versus surgical pseudotumors, Abdom. Imaging 35 (2010) 241–245, https://doi.org/10.1007/
nephrectomy for small renal tumours, Cardiovasc. Intervent. Radiol. 37 (2014) s00261-008-9499-y.
427–437, https://doi.org/10.1007/s00270-014-0846-9. [43] T.H. Oh, Y.H. Lee, I.Y. Seo, Diagnostic efficacy of contrast-enhanced ultrasound
[20] M.E. Krokidis, F. Orsi, K. Katsanos, T. Helmberger, A. Adam, CIRSE guidelines on for small renal masses, Korean J. Urol. 55 (2014) 587–592, https://doi.org/
percutaneous ablation of small renal cell carcinoma, Cardiovasc. Intervent. 10.4111/kju.2014.55.9.587.
Radiol. 40 (2017) 177–191, https://doi.org/10.1007/s00270-016-1531-y. [44] Z.F. Xu, H.X. Xu, X.Y. Xie, G.J. Liu, Y.L. Zheng, M.D. Lu, Renal cell carcinoma and
[21] M. Borghesi, E. Brunocilla, A. Volpe, H. Dababneh, C.V. Pultrone, V. Vagnoni, renal angiomyolipoma: differential diagnosis with real-time contrast-enhanced
G. La Manna, A. Porreca, G. Martorana, R. Schiavina, Active surveillance for ultrasonography, J. Ultrasound Med. 29 (2010) 709–717, https://doi.org/
clinically localized renal tumors: An updated review of current indications and 10.7863/jum.2010.29.5.709.
clinical outcomes, Int. J. Urol. 22 (2015) 432–438, https://doi.org/10.1111/ [45] Z.F. Xu, H.X. Xu, X.Y. Xie, G.J. Liu, Y.L. Zheng, J.Y. Liang, M.D. Lu, Renal cell
iju.12734. carcinoma: real-time contrast-enhanced ultrasound findings, Abdom. Imaging 35
[22] M. Nayyar, P. Cheng, B. Desai, S. Cen, M. Desai, I. Gill, V. Duddalwar, Active (2010) 750–756, https://doi.org/10.1007/s00261-009-9583-y.
surveillance of small renal masses: a review on the role of imaging with a focus on [46] M. Atri, L. Tabatabaeifar, H.J. Jang, A. Finelli, H. Moshonov, M. Jewett, Accuracy
growth rate, J. Comput. Assist. Tomogr. 40 (2016) 517–523, https://doi.org/ of contrast-enhanced US for differentiating benign from malignant solid small
10.1097/RCT.0000000000000407. renal masses, Radiology 276 (2015) 900–908, https://doi.org/10.1148/
[23] B. Musaddaq, T. Musaddaq, A. Gupta, S. Ilyas, C. von Stempel, Renal cell radiol.2015140907.
carcinoma: the evolving role of imaging in the 21st century, Semin. Ultrasound [47] D.A. Clevert, N. Minaifar, S. Weckbach, E.M. Jung, K. Stock, M. Reiser,
CT MR 41 (2020) 344–350, https://doi.org/10.1053/j.sult.2020.05.002. M. Staehler, Multislice computed tomography versus contrast-enhanced
[24] R. Kaur, M. Juneja, A.K. Mandal, An overview of non-invasive imaging modalities ultrasound in evaluation of complex cystic renal masses using the Bosniak
for diagnosis of solid and cystic renal lesions, Med. Biol. Eng. Comput. 58 (2020) classification system, Clin. Hemorheol. Microcirc. 39 (2008) 171–178.
1–24, https://doi.org/10.1007/s11517-019-02049-z. [48] B.K. Park, B. Kim, S.H. Kim, K. Ko, H.M. Lee, H.Y. Choi, Assessment of cystic renal
[25] P. Pavlica, L. Derchi, G. Martorana, E. Brunocilla, A. Bertaccini, F. Manferrari, masses based on Bosniak classification: comparison of CT and contrast-enhanced
A. Franceschelli, L. Barozzi, Renal cell carcinoma imaging, Eur. Urol. Sup. 5 US, Eur. J. Radiol. 61 (2007) 310–314, https://doi.org/10.1016/j.
(2006) 580–592, https://doi.org/10.1016/j.eursup.2006.03.010. ejrad.2006.10.004.
[26] M. Bertolotto, S. Bucci, M. Valentino, F. Currò, C. Sachs, M.A. Cova, Contrast- [49] G. Ascenti, S. Mazziotti, G. Zimbaro, N. Settineri, C. Magno, D. Melloni, R. Caruso,
enhanced ultrasound for characterizing renal masses, Eur. J. Radiol. 105 (2018) E. Scribano, Complex cystic renal masses: characterization with contrast-
41–48, https://doi.org/10.1016/j.ejrad.2018.05.015. enhanced US, Radiology 243 (2007) 158–165, https://doi.org/10.1148/
[27] M. Siddaiah, S. Krishna, M.D.F. McInnes, J.S. Quon, W.M. Shabana, D. Papadatos, radiol.2431051924.
N. Schieda, Is Ultrasound Useful for Further Evaluation of Homogeneously [50] E. Quaia, M. Bertolotto, V. Cioffi, A. Rossi, E. Baratella, R. Pizzolato, M.A. Cova,
Hyperattenuating Renal Lesions Detected on CT? AJR Am. J. Roentgenol. 209 Comparison of contrast-enhanced sonography with unenhanced sonography and
(2017) 604–610, https://doi.org/10.2214/AJR.17.17814. contrast-enhanced CT in the diagnosis of malignancy in complex cystic renal
[28] C.M. Burgan, R. Rupan Sanyal, M.E. Lockhart, Ultrasound of renal masses, Radiol. masses, AJR Am. J. Roentgenol. 191 (2008) 1239–1249, https://doi.org/
Clin. North Am. 57 (2019) 585–600, https://doi.org/10.1016/j.rcl.2019.01.009. 10.2214/AJR.07.3546.
[29] P.S. Sidhu, V. Cantisani, C.F. Dietrich, O.H. Gilja, A. Saftoiu, E. Bartels, [51] O. Graumann, S.S. Osther, J. Karstoft, A. Horlyck, P.J. Osther, Bosniak
M. Bertolotto, F. Calliada, D. Clevert, D. Cosgrove, A. Deganello, M. D’Onofrio, F. classification system: a prospective comparison of CT, contrast-enhanced US, and
M. Drudi, S. Freeman, C. Harvey, C. Jenssen, E. Jung, A.S. Klauser, N. Lassau, M. MR for categorizing complex renal cystic masses, Acta Radiol. 57 (2016)
F. Meloni, E. Leen, C. Nicolau, C. Nolsoe, F. Piscaglia, F. Prada, H. Prosch, 1409–1417, https://doi.org/10.1177/0284185115588124.
M. Radzina, L. Savelli, H. Weskott, H. Wijkstra, The EFSUMB Guidelines and [52] E.H. Chang, W.K. Chong, S.K. Kasoji, J.R. Fielding, E. Altun, L.B. Mullin, J.I. Kim,
Recommendations for the Clinical Practice of Contrast-Enhanced Ultrasound J.P. Fine, P.A. Dayton, W.K. Rathmell, Diagnostic accuracy of contrast-enhanced
(CEUS) in Non-Hepatic Applications: Update 2017 (Long Version), Ultraschall ultrasound for characterization of kidney lesions in patients with and without
Med. 39 (2018) e2–e44, https://doi.org/10.1055/s-0044-101254. chronic kidney disease, BMC Nephrol. 18 (2017) 266, https://doi.org/10.1186/
[30] T.J. van Oostenbrugge, J.J. Fütterer, P.F.A. Mulders, Diagnostic imaging for solid s12882-017-0681-8.
renal tumors: a pictorial review, Kidney Cancer 2 (2018) 79–93, https://doi.org/ [53] M. Gulati, K.G. King, I.S. Gill, V. Pham, E. Grant, V.A. Duddalwar, Contrast-
10.3233/KCA-180028. enhanced ultrasound (CEUS) of cystic and solid renal lesions: a review, Abdom.
[31] C. Lopes Vendrami, C.P. Villavicencio, T.J. DeJulio, A. Chatterjee, D.D. Casalino, Imaging 40 (2015) 1982–1996, https://doi.org/10.1007/s00261-015-0348-5.
J.M. Horowitz, D.T. Oberlin, G. Yang, P. Nikolaidis, F.H. Miller, Differentiation of [54] M. Bertolotto, C. Cicero, R. Perrone, F. Degrassi, F. Cacciato, M.A. Cova, Renal
solid renal tumors with Multiparametric MR imaging, Radiographics 37 (2017) masses with equivocal enhancement at CT: characterization with contrast-
2026–2042, https://doi.org/10.1148/rg.2017170039. enhanced ultrasound, AJR Am. J. Roentgenol. 204 (2015) W557–W565, https://
[32] F. Cornelis, N. Grenier, Multiparametric magnetic resonance imaging of solid doi.org/10.2214/AJR.14.13375.
renal tumors: a practical algorithm, Semin. Ultrasound CT MR 38 (2017) 47–58, [55] R.G. Barr, C. Peterson, A. Hindi, Evaluation of indeterminate renal masses with
https://doi.org/10.1053/j.sult.2016.08.009. contrast-enhanced US: a diagnostic performance study, Radiology 271 (2013)
[33] L. Marconi, S. Dabestani, T.B. Lam, F. Hofmann, F. Stewart, J. Norrie, A. Bex, 133–142, https://doi.org/10.1148/radiol.13130161.
K. Bensalah, S.E. Canfield, M. Hora, M.A. Kuczyk, A.S. Merseburger, P.F. [56] C.J. Harvey, A. Alsafi, S. Kuzmich, A. Ngo, I. Papadopoulou, A. Lakhani,
A. Mulders, T. Powles, M. Staehler, B. Ljungberg, A. Volpe, Systematic review and Y. Berkowitz, S. Moser, P.S. Sidhu, D.O. Cosgrove, Role of US contrast agents in
meta-analysis of diagnostic accuracy of percutaneous renal tumour biopsy, Eur. the assessment of indeterminate solid and cystic lesions in native and transplant
Urol. 69 (2016) 660–673, https://doi.org/10.1016/j.eururo.2015.07.072. kidneys, Radiographics 35 (2015) 1419–1430, https://doi.org/10.1148/
[34] N. Shi, F. Zu, Y. Shan, S. Chen, B. Xu, M. Du, M. Chen, The value of renal score in rg.2015140222.
both determining surgical strategies and predicting complications for renal cell

11
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

[57] Q. Li, Z. Wang, X. Ma, J. Tang, Y. Luo, Diagnostic accuracy of contrast-enhanced [78] G.M. Israel, M.A. Bosniak, Pitfalls in renal mass evaluation and how to avoid
ultrasound for detecting bland thrombus from inferior vena cava tumor thrombus them, Radiographics 28 (2008) 1325–1338, https://doi.org/10.1148/
in patients with renal cell carcinoma, Int. Braz. J. Urol. 46 (2020) 92–100, rg.285075744.
https://doi.org/10.1590/S1677-5538.IBJU.2019.0304. [79] K.Z. Sheir, M. El-Azab, A. Mosbah, M. El-Baz, A.A. Shaaban, Differentiation of
[58] M.F. Meloni, M. Bertolotto, C. Alberzoni, S. Lazzaroni, C. Filice, T. Livraghi, renal cell carcinoma subtypes by multislice computerized tomography, J. Urol.
G. Ferraioli, Follow-up after percutaneous radiofrequency ablation of renal cell 174 (2005) 451–455, https://doi.org/10.1097/01.ju.0000165341.08396.a9.
carcinoma: contrast-enhanced sonography versus contrast-enhanced CT or MRI, [80] J.R. Young, D. Margolis, S. Sauk, A.J. Pantuck, J. Sayre, S.S. Raman, Clear cell
AJR Am. J. Roentgenol. 191 (2008) 1233–1238, https://doi.org/10.2214/ renal cell carcinoma: discrimination from other renal cell carcinoma subtypes and
AJR.07.3238. oncocytoma at multiphasic multidetector CT, Radiology 267 (2013) 444–453,
[59] S.C. Tenant, C.M. Gutteridge, The clinical use of contrast-enhanced ultrasound in https://doi.org/10.1148/radiol.13112617.
the kidney, Ultrasound 24 (2016) 94–103, https://doi.org/10.1177/ [81] J.K. Kim, S.Y. Park, J.H. Shon, K.S. Cho, Angiomyolipoma with minimal fat:
1742271X15627185. differentiation from renal cell carcinoma at biphasic helical CT, Radiology 230
[60] M.L. Schnitzer, L. Sabel, V. Schwarze, C. Marschner, M.F. Froelich, P. Nuhn, (2004) 677–684, https://doi.org/10.1148/radiol.2303030003.
Y. Falck, M.M. Nuhn, S. Afat, M. Staehler, J. Rückel, D.-A. Clevert, J. Rübenthaler, [82] A.B. Rosenkrantz, N. Hindman, E.F. Fitzgerald, B.E. Niver, J. Melamed, J.S. Babb,
T. Geyer, Structured reporting in the characterization of renal cysts by contrast- MRI features of renal oncocytoma and chromophobe renal cell carcinoma, AJR
enhanced ultrasound (CEUS) using the bosniak classification system- Am. J. Roentgenol. 195 (2010) W421–W427, https://doi.org/10.2214/
improvement of report quality and interdisciplinary communication, Diagnostics AJR.10.4718.
(Basel) 11 (2021) 313, https://doi.org/10.3390/diagnostics11020313. [83] S. Choudhary, A. Rajesh, N.J. Mayer, K.A. Mulcahy, A. Haroon, Renal
[61] V. Schwarze, J. Rübenthaler, C. Marschner, M.P. Fabritius, J. Rueckel, N. Fink, oncocytoma: CT features cannot reliably distinguish oncocytoma from other renal
D. Puhr-Westerheide, E. Gresser, M.F. Froelich, M.L. Schnitzer, N. Große Hokamp, neoplasms, Clin. Radiol. 64 (2009) 517–522, https://doi.org/10.1016/j.
S. Afat, M. Staehler, T. Geyer, D.-A. Clevert, Advanced fusion imaging and crad.2008.12.011.
contrast-enhanced imaging (CT/MRI–CEUS) in oncology, Cancers 12 (2020) [84] G. Gakis, U. Kramer, D. Schilling, S. Kruck, A. Stenzl, H.P. Schlemmer, Small renal
2821, https://doi.org/10.3390/cancers12102821. oncocytomas: differentiation with multiphase CT, Eur. J. Radiol. 80 (2011)
[62] P. Frinking, T. Segers, Y. Luan, F.C. Tranquart, Three decades of ultrasound 274–278, https://doi.org/10.1016/j.ejrad.2010.06.049.
contrast agents: a review of the past, present and future improvements, [85] D. Giambelluca, S. Pellegrino, M. Midiri, G. Salvaggio, The "central stellate scar"
Ultrasound Med. Biol. 46 (2020) 892–908, https://doi.org/10.1016/j. sign in renal oncocytoma, Abdom. Radiol. (NY) 44 (2019) 1942–1943, https://
ultrasmedbio.2019.12.008. doi.org/10.1007/s00261-019-01899-3.
[63] K. Sasaguri, N. Takahashi, CT and MR imaging for solid renal mass [86] J.I. Kim, J.Y. Cho, K.C. Moon, H.J. Lee, S.H. Kim, Segmental enhancement
characterization, Eur. J. Radiol. 99 (2018) 40–54, https://doi.org/10.1016/j. inversion at biphasic multidetector CT: characteristic finding of small renal
ejrad.2017.12.008. oncocytoma, Radiol 252 (2009) 441–448, https://doi.org/10.1148/
[64] Z.J. Wang, A.C. Westphalen, R.J. Zagoria, CT and MRI of small renal masses, Br. radiol.2522081180.
J. Radiol. 91 (2018) 20180131, https://doi.org/10.1259/bjr.20180131. [87] N. Schieda, M. Al-Subhi, T.A. Flood, M. El-Khodary, M.D.F. McInnes, Diagnostic
[65] S. Krishna, C.A. Murray, M.D. McInnes, R. Chatelain, M. Siddaiah, O. Al-Dandan, accuracy of segmental enhancement inversion for the diagnosis of renal
S. Narayanasamy, N. Schieda, CT imaging of solid renal masses: pitfalls and oncocytoma using biphasic computed tomography (CT) and multiphase contrast-
solutions, Clin. Radiol. 72 (2017) 708–721, https://doi.org/10.1016/j. enhanced magnetic resonance imaging (MRI), Eur. Radiol. 24 (2014) 2787–2794,
crad.2017.05.003. https://doi.org/10.1007/s00330-014-3310-y.
[66] Z.J. Wang, M.S. Davenport, S.G. Silverman, H. Chandarana, A. Doshi, G.M. Israel, [88] C.G. Moldovanu, B. Petresc, A. Lebovici, A. Tamas-Szora, M. Suciu, N. Crisan,
J.R. Leyendecker, I. Pedrosa, S. Raman, E.M. Remer, A.B. Shinagare, A.D. Smith, P. Medan, M.M. Buruian, Differentiation of clear cell renal cell carcinoma from
R. Vikram, CT Renal Mass Protocol v1.0: Society of Abdominal Radiology Disease other renal cell carcinoma subtypes and benign oncocytoma using quantitative
Focused Panel on Renal Cell Carcinoma, 2020 https://doi.org/ MDCT enhancement parameters, Medicina (Kaunas) 28 (2020) 569, https://doi.
abdominalradiology.org/wp-content/uploads/2020/11/RCC.CTprotocolsfinal-7- org/10.3390/medicina56110569.
15-17.pdf. [89] B.K. Park, Renal Angiomyolipoma: Radiologic Classification and Imaging
[67] B.A. Birnbaum, J.E. Jacobs, P. Ramchandani, Multiphasic renal CT: comparison of Features According to the Amount of Fat, AJR Am. J. Roentgenol. 209 (2017)
renal mass enhancement during the corticomedullary and nephrographic phases, 826–835, https://doi.org/10.2214/AJR.17.17973.
Radiology 200 (1996) 753–758, https://doi.org/10.1148/ [90] N. Takahashi, S. Leng, K. Kitajima, D. Gomez-Cardona, P. Thapa, R.E. Carter, B.
radiology.200.3.8756927. C. Leibovich, K. Sasiwimonphan, K. Sasaguri, A. Kawashima, Small (< 4 cm) renal
[68] N.D. Egbert, E.M. Caoili, R.H. Cohan, M.S. Davenport, I.R. Francis, L.P. Kunju, J. masses: differentiation of angiomyolipoma without visible fat from renal cell
H. Ellis, Differentiation of papillary renal cell carcinoma subtypes on CT and MRI, carcinoma using unenhanced and contrast-enhanced CT, AJR Am. J. Roentgenol.
AJR Am. J. Roentgenol. 201 (2013) 347–355, https://doi.org/10.2214/ 205 (2015) 1194–1202, https://doi.org/10.2214/AJR.14.14183.
AJR.12.9451. [91] S.G. Silverman, G.M. Israel, Q.D. Trinh, Incompletely characterized incidental
[69] R.D. Ward, E.M. Remer, Cystic renal masses: an imaging update, Eur. J. Radiol. renal masses: emerging data support conservative management, Radiology 275
99 (2017) 103–110, https://doi.org/10.1016/j.ejrad.2017.12.015. (2015) 28–42, https://doi.org/10.1148/radiol.14141144.
[70] S. Narayanasamy, S. Krishna, A.K. Prasad Shanbhogue, T.A. Flood, N. Sadoughi, [92] J. Salameh, M.D.F. McInnes, T.A. McGrath, G. Salameh, N. Schieda, Diagnostic
P. Sathiadoss, N. Schieda, Contemporary update on imaging of cystic renal masses accuracy of dual-energy CT for evaluation of renal masses: systematic review and
with histopathological correlation and emphasis on patient management, Clin. meta-analysis, AJR Am. J. Roentgenol. 212 (2019) W100–W105, https://doi.org/
Radiol. 74 (2019) 83–94, https://doi.org/10.1016/j.crad.2018.09.003. 10.2214/AJR.18.20527.
[71] N. Schieda, M.S. Davenport, S. Krishna, E.A. Edney, I. Pedrosa, N. Hindman, R. [93] D. Marin, D. Davis, K.R. Choudhury, B. Patel, R.T. Gupta, A. Mileto, R.C. Nelson,
H. Baroni, N.E. Curci, A. Shinagare, S.G. Silverman, Bosniak classification of Characterization of small focal renal lesions: diagnostic accuracy with single-
cystic renal masses, version 2019: a pictorial guide to clinical use, Radiographics phase contrast-enhanced dual-energy CT with material attenuation analysis
16 (2021) 200160, https://doi.org/10.1148/rg.2021200160. compared with conventional attenuation measurements, Radiology 284 (2017)
[72] E. Edney, M.S. Davenport, N. Curci, N. Schieda, S. Krishna, N. Hindman, S. 737–747, https://doi.org/10.1148/radiol.2017161872.
G. Silverman, I. Pedrosa, Bosniak classification of cystic renal masses, version [94] F.G. Mazzei, M.A. Mazzei, N.C. Squitieri, C. Pozzessere, L. Righi, A. Cirigliano,
2019: interpretation pitfalls and recommendations to avoid misclassification, S. Guerrini, D. D’Elia, M.R. Ambrosio, A. Barone, M.T. del Vecchio, L. Volterrani,
Abdom. Radiol. (NY) 23 (2021), https://doi.org/10.1007/s00261-020-02906-8. CT perfusion in the characterization of renal lesions: an added value to
[73] B.A. Birnbaum, N. Hindman, J. Lee, J.S. Babb, Renal cyst pseudoenhancement: multiphasic CT, Biomed Res. Int. 2014 (2014), 135013, https://doi.org/10.1155/
influence of multidetector CT reconstruction algorithm and scanner type in 2014/135013.
phantom model, Radiology 244 (2007) 767–775, https://doi.org/10.1148/ [95] Z. Feng, Q. Shen, Y. Li, Z. Hu, CT texture analysis: a potential tool for predicting
radiol.2443061537. the Fuhrman grade of clear-cell renal carcinoma, Cancer Imaging 19 (2019) 6,
[74] J.H. Kim, H.Y. Sun, J. Hwang, S.S. Hong, Y.J. Cho, S.W. Doo, W.J. Yang, Y. https://doi.org/10.1186/s40644-019-0195-7.
S. Song, Diagnostic accuracy of contrast-enhanced computed tomography and [96] Y. Deng, E. Soule, A. Samuel, S. Shah, E. Cui, M. Asare-Sawiri, C. Sundaram,
contrast-enhanced magnetic resonance imaging of small renal masses in real C. Lall, K. Sandrasegaran, CT texture analysis in the differentiation of major renal
practice: sensitivity and specificity according to subjective radiologic cell carcinoma subtypes and correlation with Fuhrman grade, Eur. Radiol. 29
interpretation, World J. Surg. Oncol. 12 (2016) 260, https://doi.org/10.1186/ (2019) 6922–6929, https://doi.org/10.1007/s00330-019-06260-2.
s12957-016-1017-z. [97] J. Shu, Y. Tang, J. Cui, R. Yang, X. Meng, Z. Cai, J. Zhang, W. Xu, D. Wen, H. Yin,
[75] T. Kwon, I.G. Jeong, S. Yoo, S.J. Lee, S. Hong, D. You, J.H. Hong, H. Ahn, C. Kim, Clear cell renal cell carcinoma: CT-based radiomics features for the prediction of
Role of MRI in indeterminate renal mass: diagnostic accuracy and impact on Fuhrman grade, Eur. J. Radiol. 109 (2018) 8–12, https://doi.org/10.1016/j.
clinical decision making, Int. Urol. Nephrol. 47 (2015) 585–593, https://doi.org/ ejrad.2018.10.005.
10.1007/s11255-015-0928-x. [98] A. Bhandari, M. Ibrahim, C. Sharma, R. Liong, S. Gustafson, M. Prior, CT-based
[76] J. Patel, M.S. Davenport, S. Khalatbari, R.H. Cohan, J.H. Ellis, J.F. Platt, In vivo radiomics for differentiating renal tumours: a systematic review, Abdom. Radiol.
predictors of renal cyst pseudoenhancement at 120 kVp, AJR Am. J. Roentgenol. (NY) 2 (2020), https://doi.org/10.1007/s00261-020-02832-9.
202 (2014) 336–342, https://doi.org/10.2214/AJR.13.10915. [99] M.G. Lubner, Radiomics and artificial intelligence for renal mass characterization,
[77] N.S. Patel, L. Poder, Z.J. Wang, B.M. Yeh, A. Qayyum, H. Jin, F.V. Coakley, The Radiol. Clin. North Am. 58 (2020) 995–1008, https://doi.org/10.1016/j.
characterization of small hypoattenuating renal masses on contrast-enhanced CT, rcl.2020.06.001.
Clin. Imaging 33 (2009) 295–300, https://doi.org/10.1016/j. [100] A. Finelli, N. Ismaila, B. Bro, P. Russo, Management of small renal masses:
clinimag.2008.12.002. american society of clinical oncology clinical practice guideline, J. Clin. Oncol. 35
(2017) 668–680, https://doi.org/10.1200/JOP.2016.019620.

12
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

[101] A. Vasudevan, R.J. Davies, B.A. Shannon, R.J. Cohen, Incidental renal tumours: Am. J. Roentgenol. 216 (2021) 412–420, https://doi.org/10.2214/
the frequency of benign lesions and the role of preoperative core biopsy, BJU Int. AJR.20.23656.
97 (2006) 946–949, https://doi.org/10.1111/j.1464-410X.2006.06126.x. [125] M. Dilauro, M. Quon, M.D. McInnes, M. Vakili, A. Chung, T.A. Flood, N. Schieda,
[102] A. Sanchez, A.S. Feldman, A. Ari Hakimi, Current management of small renal Comparison of contrast- enhanced multiphase renal protocol CT versus MRI for
masses, including patient selection, renal tumor biopsy, active surveillance, and diagnosis of papillary renal cell carcinoma, AJR Am. J. Roentgenol. 206 (2016)
thermal ablation, J. Clin. Oncol. 36 (2018) 3591–3600, https://doi.org/10.1200/ 319–325, https://doi.org/10.2214/AJR.15.14932.
JCO.2018.79.2341. [126] N. Schieda, R.S. Lim, M.D.F. McInnes, I. Thomassin, R. Renard-Penna,
[103] K. Nakashima, Y. Kitagawa, K. Izumi, A. Mizokami, T. Gabata, M. Namiki, S. Tavolaro, F.H. Cornelis, Characterization of small (<4cm) solid renal masses by
Diagnostic accuracy of pre-operative imaging findings in presumed clinical T1a computed tomography and magnetic resonance imaging: current evidence and
renal cell carcinomas, Oncol. Lett. 11 (2016) 3189–3193, https://doi.org/ further development, Diagn. Interv. Imaging 99 (2018) 443–455, https://doi.org/
10.3892/ol.2016.4330. 10.1016/j.diii.2018.03.004.
[104] A.J. Bradley, L. MacDonald, S. Whiteside, R.J. Johnson, V.A.C. Ramani, Accuracy [127] F. Cornelis, E. Tricaud, A.S. Lasserre, F. Petitpierre, J.C. Bernhard, Y. Le Bras,
of preoperative CT T staging of renal cell carcinoma: which features predict M. Yacoub, M. Bouzgarrou, A. Ravaud, N. Grenier, Routinely performed
advanced stage? Clin. Radiol. 70 (2015) 822–829, https://doi.org/10.1016/j. multiparametric magnetic resonance imaging helps to differentiate common
crad.2015.03.013. subtypes of renal tumours, Eur. Radiol. 24 (2014) 1068–1080, https://doi.org/
[105] W. Chen, L. Wang, Q. Yang, B. Liu, Y. Sun, Comparison of radiographic and 10.1007/s00330-014-3107-z.
pathologic sizes of renal tumors, Int. Braz. J. Urol. 39 (2013) 189–194, https:// [128] N.K. Ramamurthy, B. Moosavi, M.D. McInnes, T.A. Flood, N. Schieda,
doi.org/10.1590/S1677-5538.IBJU.2013.02.06. Multiparametric MRI of solid renal masses: pearls and pitfalls, Clin. Radiol. 70
[106] S.E. Lee, W.K. Lee, D.S. Kim, S.H. Doo, H.Z. Park, C.Y. Yoon, S.I. Hwang, H.J. Lee, (2015) 304–316, https://doi.org/10.1016/j.crad.2014.10.006.
G. Choe, S.K. Hong, Comparison of radiographic and pathologic sizes of renal [129] M.R. Sun, L. Ngo, E.M. Genega, M.B. Atkins, M.E. Finn, N.M. Rofsky, I. Pedrosa,
tumors, World J. Urol. 28 (2010) 263–267, https://doi.org/10.1007/s00345-010- Renal cell carcinoma: dynamic contrast-enhanced MR imaging for differentiation
0511-0. of tumor subtypes-correlation with pathologic findings, Radiology 250 (2009)
[107] C. Catalano, F. Fraioli, A. Laghi, A. Napoli, F. Pediconi, M. Danti, P. Nardis, 793–802, https://doi.org/10.1148/radiol.2503080995.
R. Passariello, High-resolution multidetector CT in the preoperative evaluation of [130] J.R. Young, H. Coy, H.J. Kim, M. Douek, P. Lo, A.J. Pantuck, S.S. Raman,
patients with renal cell carcinoma, AJR Am. J. Roentgenol. 180 (2003) Performance of relative enhancement on multiphasic MRI for the differentiation
1271–1277, https://doi.org/10.2214/ajr.180.5.1801271. of clear cell renal cell carcinoma (RCC) from papillary and chromophobe RCC
[108] H.K. Sokhi, W.Y. Mok, U. Patel, Stage T3a renal cell carcinoma: staging accuracy subtypes and oncocytoma, AJR Am. J. Roentgenol. 208 (2017) 812–819, https://
of CT for sinus fat, perinephric fat or renal vein invasion, Br. J. Radiol. 88 (2015) doi.org/10.2214/AJR.16.17152.
20140504, https://doi.org/10.1259/bjr.20140504. [131] A.B. Rosenkrantz, N.E. Wehrli, J. Melamed, S.S. Taneja, M.B. Shaikh, Renal
[109] P.J. Hallscheidt, M. Bock, G. Riedasch, I. Zuna, S.O. Schoenberg, F. Autschbach, masses measuring under 2 cm: pathologic outcomes and associations with MRI
M. Soder, G. Noeldge, Diagnostic accuracy of staging renal cell carcinomas using features, Eur. J. Radiol. 83 (2014) 1311–1316, https://doi.org/10.1016/j.
multidetector-row computed tomography and magnetic resonance imaging: a ejrad.2014.05.016.
prospective study with histopathologic correlation, J. Comput. Assist. Tomogr. 28 [132] A.M. Hotker, Y. Mazaheri, A. Wibmer, C.A. Karlo, J. Zheng, C.S. Moskowitz, S.
(2004) 333–339, https://doi.org/10.1097/00004728-200405000-00005. K. Tickoo, P. Russo, H. Hricak, O. Akin, Differentiation of clear cell renal cell
[110] P.J. Hallscheidt, C. Fink, A. Haferkamp, M. Bock, A. Luburic, I. Zuna, G. Noeldge, carcinoma from other renal cortical tumors by use of a quantitative
G. Kauffmann, Preoperative staging of renal cell carcinoma with inferior vena multiparametric MRI approach, AJR Am. J. Roentgenol. 208 (2017) W85–W91,
cava thrombus using multi- detector CT and MRI: prospective study with https://doi.org/10.2214/AJR.16.16652.
histopathological correlation, J. Comput. Assist. Tomogr. 29 (2005) 64–68, [133] C. Roy Sr, S. El Ghali, X. Buy, X.V. Lindner, H. Lang, C. Saussine, D. Jacqmin,
https://doi.org/10.1097/01.rct.0000146113.56194.6d. Significance of the pseudocapsule on MRI of renal neoplasms and its potential
[111] N. Lawrentschuk, J. Gani, R. Riordan, S. Esler, D.M. Bolton, Multidetector application for local staging: a retrospective study, AJR Am. J. Roentgenol. 184
computed tomography vs magnetic resonance imaging for defining the upper (2005) 113–120, https://doi.org/10.2214/ajr.184.1.01840113.
limit of tumour thrombus in renal cell carcinoma: a study and review, BJU Int. 96 [134] R.S. Lim, T.A. Flood, M.D.F. McInnes, L.T. Lavallee, N. Schieda, Renal
(2005) 291–295, https://doi.org/10.1111/j.1464-410X.2005.05617.x. angiomyolipoma without visible fat: can we make the diagnosis using CT and
[112] Y. Sawai, T. Kinouchi, M. Mano, N. Meguro, O. Maeda, M. Kuroda, M. Usami, MRI? Eur. Radiol. 28 (2018) 542–553, https://doi.org/10.1007/s00330-017-
Ipsilateral adrenal involvement from renal cell carcinoma: retrospective study of 4988-4.
the predictive value of computed tomography, Urology 59 (2002) 28–31, https:// [135] F.U. Kay, N.E. Canvasser, Y. Xi, D.F. Pinho, D.N. Costa, A. Diaz de Leon, G. Khatri,
doi.org/10.1016/s0090-4295(01)01480-7. J.R. Leyendecker, T. Yokoo, A.H. Lay, N. Kavoussi, E. Koseoglu, J.A. Cadeddu,
[113] G.J. Nason, A. Aslam, S.K. Giri, Predictive ability of preoperative CT scan in I. Pedrosa, Diagnostic performance and interreader agreement of a standardized
determining whether the adrenal gland is spared at radical nephrectomy, Curr. MR imaging approach in the prediction of small renal mass histology, Radiology
Urol. 9 (2016) 143–147, https://doi.org/10.1159/000442869. 287 (2018) 543–553, https://doi.org/10.1148/radiol.2018171557.
[114] S. Sheth, J.C. Scatarige, K.M. Horton, F.M. Corl, E.K. Fishman, Current concepts in [136] C. Roy, B. Sauer, V. Lindner, H. Lang, C. Saussine, D. Jacqmin, MR Imaging of
the diagnosis and management of renal cell carcinoma: role of multidetector ct papillary renal neoplasms: potential application for characterization of small
and three-dimensional CT, Radiographics 21 (2001) S237–S254, https://doi.org/ renal masses, Eur. Radiol. 17 (2007) 193–200, https://doi.org/10.1007/s00330-
10.1148/radiographics.21.suppl_1.g01oc18s237. 006-0271-9.
[115] N. Griffin, M.E. Gore, S.A. Sohaib, Imaging in metastatic renal cell carcinoma, [137] M.R. Oliva, J.N. Glickman, K.H. Zou, S.Y. Teo, K.J. Mortelé, M.S. Rocha, S.
AJR Am. J. Roentgenol. 189 (2007) 360–370, https://doi.org/10.2214/ G. Silverman, Renal cell carcinoma T1 and T2 signal intensity characteristics of
AJR.07.2077. papillary and clear cell types correlated with pathology, AJR Am. J. Roentgenol.
[116] D.M. Coll, B.R. Herts, W.J. Davros, R.G. Uzzo, A.C. Novick, Preoperative use of 3D 192 (2009) 1524–1530, https://doi.org/10.2214/AJR.08.1727.
volume rendering to demonstrate renal tumors and renal anatomy, Radiographics [138] J.J. Park, C.K. Kim, Small (< 4 cm) renal tumors with predominantly low signal
20 (2000) 431–438, https://doi.org/10.1148/radiographics.20.2.g00mc16431. intensity on T2-Weighted images: differentiation of minimal-fat angiomyolipoma
[117] F. Rocco, L.A. Cozzi, G. Cozzi, Study of the renal segmental arterial anatomy with from renal cell carcinoma, AJR Am. J. Roentgenol. 208 (2017) 124–130, https://
contrast-enhanced multi-detector computed tomography, Surg. Radiol. Anat. 37 doi.org/10.2214/AJR.16.16102.
(2015) 517–526, https://doi.org/10.1007/s00276-014-1382-7. [139] C. Galmiche, J.C. Bernhard, M. Yacoub, A. Ravaud, N. Grenier, F. Cornelis, Is
[118] S. Donat, M. Diaz, J. Bishoff, J.A. Coleman, P. Dahm, I.H. Derweesh, S. Duke Multiparametric MRI Useful for Differentiating Oncocytomas From Chromophobe
Herrell 3rd, S. Hilton, E. Jonasch, D.W. Lin, V.E. Reuter, S.S. Chang, Follow-up for Renal Cell Carcinomas? AJR Am. J. Roentgenol. 208 (2017) 343–350, https://doi.
clinically localized renal neoplasms AUA guideline, J. Urol. 190 (2013) 407–416, org/10.2214/AJR.16.16832.
https://doi.org/10.1016/j.juro.2013.04.121. [140] E.A. Lassel, R. Rao, C. Schwenke, S.O. Schoenberg, H.J. Michaely, Diffusion-
[119] ESUR Guidelines on Contrast Agents v.10, European Society of Urogenital weighted imaging of focal renal lesions: a meta-analysis, Eur. Radiol. 24 (2014)
Radiology, 2021. www.esur-cm.org. 241–249, https://doi.org/10.1007/s00330-013-3004-x.
[120] Z.J. Wang, M.S. Davenport, S.G. Silverman, H. Chandarana, A. Doshi, G.M. Israel, [141] M. Tordjman, R. Mali, G. Madelin, V. Prabhu, S.K. Kang, Diagnostic test accuracy
J.R. Leyendecker, I. Pedrosa, S. Raman, E.M. Remer, A.B. Shinagare, A.D. Smith, of ADC values for identification of clear cell renal cell carcinoma: systematic
R. Vikram, MRI Renal Mass Protocol v1.0: Society of Abdominal Radiology review and meta-analysis, Eur. Radiol. 30 (2020) 4023–4038, https://doi.org/
Disease Focused Panel on Renal Cell Carcinoma, 2020 https://doi.org/ 10.1007/s00330-020-06740-w.
abdominalradiology.org/wp-content/uploads/2020/11/RCC.MRIprotocolfinal-7- [142] A. Goyal, R. Sharma, A.S. Bhalla, S. Gamanagatti, A. Seth, V.K. Iyer, P. Das,
15-17.pdf. Diffusion-weighted MRI in renal cell carcinoma: a surrogate marker for predicting
[121] G.M. Israel, N. Hindman, M.A. Bosniak, Evaluation of cystic renal masses: nuclear grade and histological subtype, Acta Radiol. 53 (2012) 349–358, https://
comparison of CT and MR imaging by using the Bosniak classification system, doi.org/10.1258/ar.2011.110415.
Radiology 231 (2004) 365–371, https://doi.org/10.1148/radiol.2312031025. [143] X. Yu, M. Lin, H. Ouyang, C. Zhou, H. Zhang, Application of ADC measurement in
[122] A.B. Rosenkrantz, N.E. Wehrli, T.C. Mussi, S.S. Taneja, M.J. Triolo, Complex characterization of renal cell carcinomas with different pathological types and
cystic renal masses: comparison of cyst complexity and Bosniak classification grades by 3.0T diffusion-weighted MRI, Eur. J. Radiol. 81 (2012) 3061–3066,
between 1.5 T and 3 T MRI, Eur. J. Radiol. 83 (2014) 503–508, https://doi.org/ https://doi.org/10.1016/j.ejrad.2012.04.028.
10.1016/j.ejrad.2013.11.013. [144] E. Inci, E. Hocaoglu, S. Aydin, T. Cimilli, Diffusion-weighted magnetic resonance
[123] N.M. Hindman, Imaging of cystic renal masses, Urol. Clin. North Am. 45 (2018) imaging in evaluation of primary solid and cystic renal masses using the Bosniak
331–349, https://doi.org/10.1016/j.ucl.2018.03.006. classification, Eur. J. Radiol. 81 (2012) 815–820, https://doi.org/10.1016/j.
[124] J.R. Tse, J. Shen, L. Shen, L. Yoon, A. Kamaya, Bosniak classification of cystic ejrad.2011.02.024.
renal masses version 2019: comparison of categorization using CT and MRI, AJR

13
A.C. Tsili et al. European Journal of Radiology 141 (2021) 109777

[145] S. Sevcenco, G. Heinz-Peer, L. Ponhold, D. Javor, F.E. Kuehhas, H.C. Klingler, Radiofrequency ablation for renal Cancer in von hippel-lindau syndrome patients:
M. Remzi, P. Weibl, S.F. Shariat, P.A. Baltzer, Utility and limitations of 3-Tesla a prospective cohort analysis, Clin. Genitourin. Cancer 10 (2017), https://doi.
diffusion-weighted magnetic resonance imaging for differentiation of renal org/10.1016/j.clgc.2017.07.027. S1558-7673(17)30237-30239.
tumors, Eur. J. Radiol. 83 (2014) 909–913, https://doi.org/10.1016/j. [166] M. Krokidis, S. Spiliopoulos, M. Jarzabek, N. Fotiadis, T. Sabharwal, T. O’Brien,
ejrad.2014.02.026. A. Adam, Percutaneous radiofrequency ablation of small renal tumours in patients
[146] K.M. Krajewski, A.B. Shinagare, Novel imaging in renal cell carcinoma, Curr. with a single functioning kidney: long-term results, Eur. Radiol. 23 (2013)
Opin. Urol. 26 (2016) 388–395, https://doi.org/10.1097/ 1933–1939, https://doi.org/10.1007/s00330-013-2802-5.
MOU.0000000000000314. [167] R. Hoffmann, T.F. Jakobs, C.H. Kubisch, C. Trumm, C. Weber, M. Siebels, T.
[147] Y. Ding, Q. Tan, W. Mao, D. Chenchen, H. Xiaoyi, H. Jun, Z. Mengsu, Z. Jianjun, K. Helmberger, M.F. Reiser, Renal cell carcinoma in patients with a solitary
Differentiating between malignant and benign renal tumors: do IVIM and kidney after nephrectomy treated with radiofrequency ablation: mid term results,
diffusion kurtosis imaging perform better than DWI? Eur. Radiol. 29 (2019) Eur. J. Radiol. 73 (2010) 652–656, https://doi.org/10.1016/j.ejrad.2008.12.015.
6930–6939, https://doi.org/10.1007/s00330-019-06240-6. [168] S. Mylona, A. Kokkinaki, M. Pomoni, P. Galani, S. Ntai, L. Thanos, Percutaneous
[148] A. Goyal, A. Razik, D. Kandasamy, A. Seth, P. Das, B. Ganeshan, R. Sharma, Role radiofrequency ablation of renal cell carcinomas in patients with solitary kidney:
of MR texture analysis in histological subtyping and grading of renal cell 6 years experience, Eur. J. Radiol. 69 (2009) 351–356, https://doi.org/10.1016/j.
carcinoma: a preliminary study, Abdom. Radiol. (NY) 44 (2019) 3336–3349, ejrad.2007.10.004.
https://doi.org/10.1007/s00261-019-02122-z. [169] A. Uhlig, O. Hahn, A. Strauss, J. Lotz, L. Trojan, R. Müller-Wille, J. Uhlig,
[149] J.H. Min, C.K. Kim, B.K. Park, E. Kim, B. Kim, Assessment of renal lesions with Treatment for localized T1a clear cell renal cell carcinoma: survival benefit for
blood oxygenation level dependent MRI at 3 T: preliminary experience, AJR Am. cryosurgery and thermal ablation compared to deferred therapy, Cardiovasc.
J. Roentgenol. 197 (2011) W489–W494, https://doi.org/10.2214/AJR.10.6319. Intervent. Radiol. 41 (2018) 277–283, https://doi.org/10.1007/s00270-017-
[150] W. Wang, K. Cao, S. Jin, X. Zhu, J. Ding, W. Peng, Differentiation of renal cell 1816-9.
carcinoma subtypes through MRI-based radiomics analysis, Eur. Radiol. 57 [170] R. El Dib, N.J. Touma, A. Kapoor, Cryoablation vs radiofrequency ablation for the
(2020) 5738–5747, https://doi.org/10.1007/s00330-020-06896-5. treatment of renal cell carcinoma: a meta-analysis of case series studies, BJU Int.
[151] E. Cui, Z. Li, C. Ma, Q. Li, Y. Lei, Y. Lan, J. Yu, Z. Zhou, R. Li, W. Long, F. Lin, 110 (2012) 510–516, https://doi.org/10.1111/j.1464-410X.2011.10885.x.
Predicting the ISUP grade of clear cell renal cell carcinoma with multiparametric [171] D.A. Kunkle, R.G. Uzzo, Cryoablation or radiofrequency ablation of the small
MR and multiphase CT radiomics, Eur. Radiol. 30 (2020) 2912–2921, https://doi. renal mass: a meta-analysis, Cancer 113 (2008) 2671–2680, https://doi.org/
org/10.1007/s00330-019-06601-1. 10.1002/cncr.23896.
[152] H.A. Ali, M.J. Couch, R. Menezes, A.J. Evans, A. Finelli, M.A. Jewett, K.S. Jhaveri, [172] T.D. Atwell, G.D. Schmit, S.A. Boorjian, J. Mandrekar, A. Nicholas Kurup, A.
Predictive value of in vivo MR spectroscopy with semilocalization by adiabatic J. Weisbrod, G.K. Chow, B.C. Leibovich, M.R. Callstrom, D.E. Patterson, C.
selective refocusing in differentiating clear cell renal cell carcinoma from other M. Lohse, R. Houston Thompson, Percutaneous ablation of renal masses
subtypes, AJR Am. J. Roentgenol. 214 (2020) 817–824, https://doi.org/10.2214/ measuring 3.0 cm and smaller: comparative local control and complications after
AJR.19.22023. radiofrequency ablation and cryoablation, AJR Am. J. Roentgenol. 200 (2013)
[153] R. Papalia, V. Panebianco, R. Mastroianni, M. Del Monte, E. Altobelli, E. Faiella, 461–466, https://doi.org/10.2214/AJR.12.8618.
F.R. Grasso, M. Bellangino, G. Simone, M. Ciccozzi, S. Angeletti, G. D’ovidio, [173] N.J. Hegarty, I.S. Gill, M.M. Desai, E.M. Remer, C.M. O’Malley, J.H. Kaouk,
C. Catalano, M. Gallucci, R.M. Scarpa, G. Muto, Accuracy of magnetic resonance Probe-ablative nephron-sparing surgery: cryoablation versus radiofrequency
imaging to identify pseudocapsule invasion in renal tumors, World J. Urol. 38 ablation, Urology 68 (2006) 7–13, https://doi.org/10.1016/j.
(2020) 407–415, https://doi.org/10.1007/s00345-019-02755-1. urology.2005.12.049.
[154] L.C. Adams, B. Ralla, Y.Y. Bender, K. Bressem, B. Hamm, J. Busch, F. Fuller, M. [174] A.J. Gunn, N.S. Parikh, S. Bhatia, Society of interventional radiology quality
R. Makowski, Renal cell carcinoma with venous extension: prediction of inferior improvement standards on percutaneous ablation in renal cell carcinoma, J. Vasc.
vena cava invasion by MRI, Cancer Imaging 18 (2018) 17, https://doi.org/ Interv. Radiol. 31 (2020) 195–201, https://doi.org/10.1016/j.jvir.2019.11.004.
10.1186/s40644-018-0150-z. [175] I.J. Patel, S. Rahim, J.C. Davidson, S.E. Hanks, A.L. Tam, T. Gregory Walker, L.
[155] M.E. Krokidis, P. Kitrou, S. Spiliopoulos, K. Karnabatidis, K. Katsanos, Image- R. Wilkins, R. Sarode, I. Weinberg, Society of Interventional Radiology Consensus
guided minimally invasive treatment for small renal cell carcinoma, Insights Guidelines for the Periprocedural management of thrombotic and bleeding risk in
Imaging 9 (2018) 385–390, https://doi.org/10.1007/s13244-018-0607-4. patients undergoing percutaneous image-guided interventions. Part II:
[156] M. Rossi, G. Orgera, A. Hatzidakis, M. Krokidis, Minimally invasive ablation recommendations: endorsed by the Canadian Association for Interventional
treatment for locally advanced pancreatic adenocarcinoma, Cardiovasc. Radiology and the Cardiovascular and Interventional Radiological Society of
Intervent. Radiol. 37 (2014) 586–591, https://doi.org/10.1007/s00270-013- Europe, J. Vasc. Interv. Radiol. 30 (2019) 1168–1184, https://doi.org/10.1016/j.
0724-x. jvir.2019.04.017.
[157] D.A. Gervais, F.J. McGovern, B.J. Wood, S.N. Goldberg, W.S. McDougal, P. [176] M. Krokidis, C. Tappero, D. Bogdanovic, K. Ziebarth, A.C. Stamm, Computed
R. Mueller, Radio-frequency ablation of renal cell carcinoma: early clinical tomography guided navigation assisted percutaneous ablation of osteoid osteoma
experience, Radiology 217 (2000) 665–672, https://doi.org/10.1148/ in a 7-year-old patient: the low dose approach, Skeletal Radiol. 46 (2017)
radiology.217.3.r00dc39665. 989–993, https://doi.org/10.1007/s00256-017-2620-y.
[158] L.W. Organ, Electrophysiologic principles of radiofrequency lesion making, Appl. [177] A. Babaei Jandaghi, M. Lebady, A.A. Zamani, A. Heidarzadeh, A. Monfared,
Neurophysiol. 39 (1976) 69–76, https://doi.org/10.1159/000102478 (1976- R. Pourghorban, A randomised clinical trial to compare coaxial and noncoaxial
1977). techniques in percutaneous core needle biopsy of renal parenchyma, Cardiovasc.
[159] S.P. Psutka, A.S. Feldman, W.S. McDougal, F.J. McGovern, P. Mueller, D. Intervent. Radiol. 40 (2017) 106–111, https://doi.org/10.1007/s00270-016-
A. Gervais, Long-term oncologic outcomes after radiofrequency ablation for T1 1466-3.
renal cell carcinoma, Eur. Urol. 63 (2013) 486–492, https://doi.org/10.1016/j. [178] J. Garnon, R.L. Cazzato, J. Caudrelier, M. Nouri-Neuville, P. Rao, E. Boatta,
eururo.2012.08.062. N. Ramamurthy, G. Koch, A. Gangi, Adjunctive thermoprotection during
[160] B. Feng, P. Liang, Local thermal ablation of renal cell carcinoma, Eur. J. Radiol. percutaneous thermal ablation procedures: review of current techniques,
81 (2012) 437–440, https://doi.org/10.1016/j.ejrad.2010.12.056. Cardiovasc. Intervent. Radiol. 42 (2019) 344–357, https://doi.org/10.1007/
[161] N.E. Hoffmann, J.C. Bischof, The cryobiology of cryosurgical injury, Urology 60 s00270-018-2089-7.
(2002) 40–49, https://doi.org/10.1016/s0090-4295(02)01683-7. [179] X. Li, P. Liang, J. Yu, Y. Xiao-Ling, L. Fang-Yi, C. Zhi-Gang, H. Zhi-Yu, Role of
[162] C.S. Georgiades, R. Rodriguez, Efficacy and safety of percutaneous cryoablation contrast-enhanced ultrasound in evaluating the efficiency of ultrasound guided
for stage 1A/B renal cell carcinoma: results of a prospective, single-arm, 5-year percutaneous microwave ablation in patients with renal cell carcinoma, Radiol.
study, Cardiovasc. Intervent. Radiol. 37 (2014) 1494–1499, https://doi.org/ Oncol. 47 (2013) 398–404, https://doi.org/10.2478/raon-2013-0038.
10.1007/s00270-013-0831-8. [180] H.J. Lee, H.J. Chung, H.K. Wang, S.H. Shen, Y.H. Chang, C.K. Chen, H.P. Chou, Y.
[163] T. Kawabata, T. Hiraki, T. Iguchi, Y. Matsui, M. Uka, Y. Masaoka, T. Komaki, Y. Chiou, Evolutionary magnetic resonance appearance of renal cell carcinoma
J. Sakurai, H. Gobara, M. Araki, Y. Nasu, S. Kanazawa, Post-ablation syndrome after percutaneous cryoablation, Br. J. Radiol. 89 (2016) 20160151, https://doi.
after percutaneous cryoablation of small renal tumors: a prospective study of org/10.1259/bjr.20160151.
incidence, severity, duration, and effect on lifestyle, Eur. J. Radiol. 122 (2020) [181] C.J. Weight, J.H. Kaouk, N.J. Hegarty, E.M. Remer, C.M. O’Malley, B.R. Lane, I.
108750, https://doi.org/10.1016/j.ejrad.2019.108750. S. Gill, A.C. Novick, Correlation of radiographic imaging and histopathology
[164] J. Yu, G. Zhang, P. Liang, Xl. Yu, Z. Cheng, Z. Han, X. Zhang, J. Dong, Q. Li, following cryoablation and radio frequency ablation for renal tumors, J. Urol. 179
M. Mu, X. Li, Midterm results of percutaneous microwave ablation under (2008) 1277–1281, https://doi.org/10.1016/j.juro.2007.11.075.
ultrasound guidance versus retroperitoneal laparoscopic radial nephrectomy for [182] T.M. Wah, H.C. Irving, W. Gregory, J. Cartledge, A.D. Joyce, P.J. Selby,
small renal cell carcinoma, Abdom. Imaging 40 (2015) 3248–3256, https://doi. Radiofrequency ablation (RFA) of renal cell carcinoma (RCC): experience in 200
org/10.1007/s00261-015-0500-2. tumours, BJU Int. 113 (2014) 416–428, https://doi.org/10.1111/bju.12349.
[165] M. Allasia, F. Soria, A. Battaglia, C. Gazzera, M. Calandri, M.P. Caprino,
B. Lucatello, A. Velrti, M. Maccario, B. Pasini, A. Bosio, P. Gontero, P. Destefanis,

14

You might also like