Retroperitoneal Tumors



Fig. 6.1
Schematic diagram of retroperitoneal anatomy at the level of the kidneys. The retroperitoneum is subdivided into the three distinct compartments: the anterior pararenal (APS), the perirenal (PRS), and the posterior pararenal (PPS) spaces. A aorta, V inferior vena cava, P pancreas, S spleen, L liver, C colon, RK right kidney, LK left kidney, PP parietal peritoneum, ARF anterior renal fascia, LCF lateroconal fascia, PRF posterior renal fascia, TF transversalis fascia



Primary retroperitoneal tumors present a broad spectrum of pathologic entities and arise from actual tissues in the same space or from embryonic rests found therein [4]. Diagnosis of these tumors begins with affirmation of their retroperitoneal location and then determination of whether the lesion is primarily retroperitoneal or is arising secondarily from a retroperitoneal organ [5]. Primary retroperitoneal tumors can be further categorized into four main groups: mesodermal neoplasms; neurogenic tumors; germ cell, sex cord, and stromal tumors; and lymphoid neoplasms [5] (Table 6.1). When lymph nodes are regarded as a retroperitoneal organ, lymphoma is not considered a primary retroperitoneal tumor. Because of high incidence of retroperitoneal lymphadenopathy including metastases and lymphoma, the term primary retroperitoneal tumors should be considered after exclusion of lymphadenopathy and anatomic connection to any retroperitoneal organs.


Table 6.1
Classification of primary retroperitoneal tumors
































Tissue of origin

Benign

Malignant

Mesodermal origin

 Adipocytic

 Smooth muscle

 Skeletal muscle

 Fibroblastic

 Vascular

Lipoma

Leiomyoma

Rhabdomyoma

Fibroma

Hemangioma

Lymphangioma

Liposarcoma

Leiomyosarcoma

Rhabdomyosarcoma

Fibrosarcoma

Hemangioendothelioma

Nervous origin

 Nerve sheath

 Sympathetic nervous system

 Chromaffin/paraganglionic cells

Schwannoma

Neurofibroma

Ganglioneuroma

Paraganglioma

Malignant peripheral nerve sheath tumor

Ganglioneuroblastoma

Neuroblastoma

Malignant paraganglioma

Germ cell, sex cord, and stromal cell origin

 Embryonic rests

 Notochord

 Embryonic hindgut

Benign teratoma

Chordoma

Tailgut cyst (retrorectal cystic hamartoma)

Malignant teratoma

Lymphoid origin
 
Lymphoma

Miscellaneous

Erdheim–Chester disease

Castleman’s disease
 

Malignant retroperitoneal tumors account for approximately 0.1 % of all malignancies and are more common than benign tumors in the retroperitoneal space [6, 7]. Retroperitoneal sarcomas account for 90 % of mesodermal primary retroperitoneal malignancies, with liposarcoma, leiomyosarcoma, and malignant fibrous histiocytoma making up more than 80 % of these tumors [1, 8]. Retroperitoneal sarcomas have a peak incidence in the fifth and sixth decades of life and occur with equal frequency in males and females [8].

They usually reach a large size without producing symptoms. Therefore, they can present later in life with nonspecific symptoms such as abdominal pain and fullness and usually have poor prognosis. The 5-year survival rate of the patients including those with sarcoma was reportedly 22–50 % [9]. Therefore, it is important to diagnose retroperitoneal tumors as early as possible and discriminate the malignant from the benign tumors precisely in order to properly treat and to avoid unnecessary aggressive treatment for benign tumors. However, since the diagnostic imaging findings of these tumors are not specific for the histological diagnosis and even imaging-guided biopsy of large retroperitoneal tumors may not be conclusive, prediction of the definitive histological diagnosis remains a challenge [7, 10].



6.2 Diagnostic Evaluation


Cross-sectional imaging plays an important role in the characterization of these masses and in the evaluation of their extent, direct involvement of adjacent structures, and detection of distant metastases and therefore in treatment planning. It is also useful in guiding biopsy, planning surgery, evaluating response to treatment, restaging, and in the long-term follow-up for local recurrence. An abdominopelvic computed tomography (CT) scan is the most commonly used modality for evaluating retroperitoneal disease, whereas magnetic resonance imaging (MRI) is more often used as a problem-solving tool. In most cases, CT is less sensitive to motion artifact than MRI, and it better defines the anatomic relationship of the tumor to other abdominal organs. CT provides superior spatial resolution and detection of calcification, while MRI has superior soft tissue contrast and capabilities in the detection of fat within a lesion. MRI has advantage in assessing fat or hemorrhage and is particularly useful in differentiating solid from nonenhancing cystic or necrotic lesions and in evaluating the extent of disease and the presence and nature of vascular thrombosis or encasement [11].

In order to classify a mass as primary retroperitoneal, the location should be determined as within the retroperitoneal space and an organ of origin is excluded. Displacement of normal retroperitoneal organs or large vessels in the space strongly suggests that a mass is retroperitoneal in location (Fig. 6.2) [3]. Several radiological signs have been described to assist in determining the organ of origin. They include claw or beak sign, invisible or phantom organ sign, and embedded organ sign (Fig. 6.3) [3, 12, 13]. The claw or beak sign is positive when a mass causes the edge of an adjacent organ into a beak shape, meaning that the mass originates from that organ. Conversely, if a mass originates from a structure adjacent to the organ, it will form obtuse angles to abut and compress the organ. The invisible or phantom organ sign is positive when a large mass arises from a small organ that then becomes undetectable. The embedded organ sign is positive when a mass that arises from a given organ often appears embedded within it and the interface between the two may be difficult to appreciate [3]. Conversely, a mass that abuts but does not originate from a hollow structure compresses it and produces a crescentic deformity. Rounded rather than beaked edges of an adjacent organ (negative beak sign) with a crescentic deformation (negative embedded organ sign) by the tumor suggest a primary retroperitoneal tumor [3].

A330234_1_En_6_Fig2_HTML.gif


Fig. 6.2
Determining tumor location within the retroperitoneal space. (a) Anterior displacement of retroperitoneal structures such as pancreas (arrows) in this case of schwannoma suggests that the mass arises in the retroperitoneum. (b) Anteromedial displacement of right iliac vessels (arrow) by the mass (leiomyosarcoma) suggests that the mass arises in the retroperitoneum


A330234_1_En_6_Fig3_HTML.gif


Fig. 6.3
Determining origin of the mass. (a) Positive beak sign: a large mass causes the edge of the kidney to become beak shaped (arrows), meaning that the lesion originates from the kidney and is not primary retroperitoneal tumor. The mass was confirmed as renal cell carcinoma. (b) Negative beak sign: a large low-density mass abuts left kidney with dull edge (arrows), meaning that the mass originates from adjacent to left kidney. The mass was confirmed as leiomyosarcoma. (c) Positive invisible or phantom organ sign: a heterogeneous mass (arrows) that was confirmed as leiomyosarcoma is located between the right kidney and abdominal aorta. The tumor’s originating organ (inferior vena cava) appears totally incorporated by the tumor at this level and is no longer recognizable. (d) Negative embedded organ sign: the wall of inferior vena cava is compressed extrinsically from the tumor creating a crescent shape (arrow), meaning that the lesion does not originate from the IVC. The mass was confirmed as right adrenal pheochromocytoma

Location, size, and anatomical changes secondary to tumor growth are easily visualized, and tumor invasion of adjacent organs can be demonstrated or suggested on cross-sectional imaging. Although there are significant overlaps in the imaging characteristics of retroperitoneal tumors, the radiographic appearances can offer clues as to the histologic subtype and grade, which may guide decisions. Some lesions have distinctive characteristics and can be diagnosed with some accuracy on imaging. It is also possible to narrow the differential diagnosis of a retroperitoneal mass based on certain imaging characteristics in combination with the pattern of involvement and demographics [5].

If fatty components are obvious, a diagnosis of liposarcoma is possible, but differentiation from a benign lipoma can be difficult. Cross-sectional imaging cannot reliably distinguish between retroperitoneal lymphomas and sarcomas: although lymphomas tend to be homogeneous on imaging and often envelope the inferior vena cava and aorta, while sarcomas are usually heterogeneous, these findings are unreliable. Imaging evidence of tumor necrosis suggests a high-grade component and portends a poor prognosis [14]. In general, extensive vascular involvement, peritoneal implants, and distant metastatic disease suggest unresectability.

Histological confirmation is required for diagnosis in many cases of primary retroperitoneal sarcomas because of overlap of imaging features and for tumor grading. However, when the diagnosis seems straightforward, preoperative tissue diagnosis is not necessary if surgery is planned to be the primary therapeutic intervention. Percutaneous biopsy can be a safe method in providing a tissue diagnosis for a locally advanced or unresectable lesion. Retroperitoneal biopsies can be safely performed under CT or US guidance. Imaging helps to identify and to guide specific targeted biopsy from the enhancing or soft tissue component of the mass, which is more likely to be the more aggressive component of the mass.


6.3 Tumors of Mesodermal Origin


Most of the primary retroperitoneal tumors are of mesodermal origin with liposarcoma, leiomyosarcoma, and malignant fibrous histiocytoma together, making up more than 80 % of primary retroperitoneal sarcomas [1, 15]. Historically, undifferentiated/unclassified soft tissue sarcomas are new terminology that replace or include the older term “malignant fibrous histiocytoma (MFH)” [16, 17]. The remaining primary retroperitoneal masses arise predominantly from the nervous system. Among children, the most common histologic types of retroperitoneal sarcomas are rhabdomyosarcoma and fibrosarcoma [18].

Retroperitoneal sarcomas typically present in the sixth and seventh decades of life and are often large at the time of clinical presentation because the loose connective tissue in the retroperitoneum provides little resistance to their growth [1]. Surgical resection is the only potentially curative treatment for a localized retroperitoneal sarcoma. The ability to perform a complete surgical resection at the time of initial presentation is the most important independent prognostic factor for survival followed only by histologic grade [5, 1921]. The usual reasons for unresectability are extensive vascular involvement or the presence of multiple peritoneal implants. Overall, regional lymphadenopathy is uncommon in soft tissue sarcomas, with a frequency of less than 4 % at presentation, and less than one-third of patients have metastases at presentation [21]. The recurrence rates are high, and metastases to the liver, lung, bones, and brain may be seen [8, 22].


6.3.1 Liposarcoma


Liposarcoma is the most common (33 %) sarcoma to occur in the retroperitoneum [22]. Retroperitoneal liposarcomas account for 10–15 % of all liposarcomas, and they are more common in the 50–70-year age group, with no sex predilection [1, 22]. They are classified histologically into four main subtypes: atypical lipomatous tumor/well differentiated, dedifferentiated, myxoid, and pleomorphic (Fig. 6.4) [15, 23]. Different histological subtypes may coexist in the same lesion. Such histological subtypes are usually classified on the basis of the most aggressive cellular component. The fourth edition of the World Health Organization (WHO) Classification of Tumors of Soft Tissue and Bone was published in February 2013 [16]. The changes for adipocytic tumors in the fourth edition were the removal of the terms “round cell liposarcoma” and “mixed-type liposarcoma” [16, 17]. While the section heading lists only “atypical lipomatous tumor” on the recent WHO classification, the authors point out that retention of “well-differentiated liposarcoma” is still appropriate for tumors in sites at which margin-negative resection is often impossible (e.g., retroperitoneum, mediastinum), since such tumors are associated with substantial mortality [17]. The terminology “atypical lipomatous tumor” and “well-differentiated liposarcoma” continues to need clarification [17].

A330234_1_En_6_Fig4_HTML.gif


Fig. 6.4
Liposarcoma with four histologic types on post-contrast CT images. (a) Well-differentiated liposarcoma in a 79-year-old man is shown as a large homogeneous fat-containing mass with thick septa (arrow) in right perirenal space with superior displacement of the right kidney (not shown). (b) Dedifferentiated liposarcoma in a 58-year-old man is shown as a large well-marginated mass with heterogeneous attenuation and enhancing septa and soft tissue components (arrow). (c) Myxoid liposarcoma in a 1-year-old man is shown as a heterogeneous hypoattenuating mass with ground-glass appearance. (d) Pleomorphic liposarcoma in a 58-year-old man is shown as a large solid soft tissue mass with internal necrosis and no fatty component

Liposarcoma is usually large (average diameter, >20 cm) and is a slow-growing tumor [1]. In general, liposarcomas may be distinguished based on the presence of fat, which varies depending on the subtype [1, 24]. The well-differentiated subtype is low-grade tumor and the most common type of retroperitoneal liposarcoma [1, 22]. The fact that well-differentiated liposarcoma shows no potential for metastasis unless it undergoes dedifferentiation led, in the late 1970s, to the introduction of terms such as atypical lipoma or atypical lipomatous tumor, particularly for lesions arising at surgically amenable locations in the limbs and on the trunk since, at these sites, wide excision should usually be curative, and hence the designation “sarcoma” is not warranted [25]. However, in sites such as the retroperitoneum and mediastinum, it is commonly impossible to obtain a wide surgical excision margin, and, in such cases, local recurrence is almost inevitable and often leads to death, even in the absence of dedifferentiation and metastasis – hence, at these sites, retention of the term well-differentiated liposarcoma can readily be justified [17, 26].

Well-differentiated liposarcomas are predominantly hypoattenuating lesion with fat attenuation on CT images and typically round or lobulated with smooth margin and may displace or surround local structures (Fig. 6.4a). The fat content in the mass demonstrates high signal intensity on T1-weighted images and intermediate signal intensity on T2-weighted images, with loss of signal on fat-suppressed images. The macroscopic fat content may be greater than 75 %, and these lesions must be distinguished from other fat-containing neoplasms which may occur in the retroperitoneum, such as lipoma, adrenal myelolipoma, renal angiomyolipoma, and teratoma [27]. Retroperitoneal lipomas are fat-containing lesions that do not have any associated soft tissue component or nodularity. Although the appearance of well-differentiated liposarcoma may be similar to that of a lipoma, this subtype frequently has thicker, irregular, and nodular septa that show enhancement after contrast material administration [28]. Calcification or ossification is rare and considered a poor prognostic feature, which often indicates dedifferentiation [3]. Well-differentiated tumors can recur but do not metastasize [1].

Dedifferentiated liposarcomas are high-grade tumors with poor prognosis [29]. This subtype is defined by the presence of sharply demarcated regions of non-lipogenic sarcomatous tissue within a well-differentiated tumor [16]. At CT and MR imaging, these dedifferentiated tumors appear as more heterogeneous tumors with both fat and solid components (Figs. 6.4b and 6.5) [1]. There may be no evidence of fat in up to 20 %, which makes the diagnosis difficult based on imaging alone [28]. Calcification is seen in as many as 30 % of these tumors and is an important sign of dedifferentiation [1, 30]. Variable signal intensity and enhancement of the solid portion may be seen [30].

A330234_1_En_6_Fig5_HTML.gif


Fig. 6.5
Dedifferentiated liposarcoma in a 63-year-old man. (a) Contrast-enhanced axial CT demonstrates a poorly marginated and markedly heterogeneous mass with fatty component (arrows) and enhancing nonadipose solid-appearing regions (small arrows) in left retroperitoneal space. The tumor invades into left psoas and quadratus lumborum muscles medially and displaces the left kidney and descending colon (black arrow) anteriorly. (b) T1-weighted axial image shows fat component (arrows) with hyperintensity in the mass. (c) T1-weighted fat-suppressed gadolinium-enhanced MRI demonstrates reduction of the signal intensity at the fat component (arrow) and significant enhancement at the nonadipose solid component of the mass. (d) Diffusion-weighted imaging shows hyperintense signal (arrow) within the solid component of the mass

Myxoid liposarcoma is of intermediate-grade malignancy and the second most common subtype of liposarcoma, accounting for more than one-third of liposarcomas and representing about 10 % of all adult soft tissue sarcomas. The lesions formerly known as round cell liposarcoma are interpreted as histologic continuum with myxoid subtype and are included in this category. This subtype shows a heterogeneous hypoattenuating mass, with attenuation less than that of muscle at CT (Fig. 6.4c). Homogeneous distribution of fat and soft tissue within the mass may result in a “pseudocystic” appearance, near fluid density, on non-contrast scan [15]. They characteristically show gradual, heterogeneous enhancement [15]. There is no macroscopic fat in over 50 % and calcifications are uncommon [28]. At MR imaging, there is low signal intensity on T1-weighted images and high signal intensity on T2-weighted images because of the mucopolysaccharide contents in the myxoid matrix [1]. Lacy, linear, or amorphous areas of high signal intensity on T1-weighted images and intermediate signal intensity on T2-weighted images may be seen because of the intratumoral fat content [1]. They demonstrate slowly progressive, reticular contrast enhancement due to solid components, which enables differentiation from a cyst [31].

Pleomorphic liposarcomas are high-grade sarcomas and the least common histologic subtype. This subtype tends to occur in the extremities (lower > upper limbs), whereas the trunk and the retroperitoneum are less frequently affected. They are seen as heterogeneous soft tissue masses with little or no fat within and may contain areas of necrosis that are indistinguishable from other solid tumors (Fig. 6.4d) [1]. Calcifications are rare.

Local recurrence after resection is the most common cause of relapse and is usually seen within 2 years. Recurrent liposarcomas may be difficult to differentiate from retroperitoneal fat, but recurrent tumors frequently have a slightly higher attenuation than the surrounding fat [13]. Recurrent liposarcomas often have a different appearance from the primary tumor and in many cases may not contain any visible fat.


6.3.2 Leiomyosarcoma


Leiomyosarcomas are the second most common primary retroperitoneal sarcoma, accounting for 28 % of all cases [22]. They arise from smooth muscle elements within the retroperitoneal muscle tissue, blood vessels, or Wolffian duct remnants [1, 19]. Leiomyosarcoma is more common in women, in the fifth to sixth decades of life [32]. They can grow to a large size (>10 cm) before compromising adjacent organs and precipitating clinical symptoms such as venous thrombosis [1]. Histopathologically, this tumor has large areas of necrosis and cystic degeneration, but calcification is uncommon [1]. They are large, well-circumscribed masses, which is isoattenuating to muscle on CT scan (Fig. 6.6). Contrast enhancement is often heterogeneous and predominantly peripheral. Small tumors may be homogeneously solid, but large tumors have extensive areas of low attenuation and represent necrosis and cystic degeneration. Rarely, leiomyosarcoma may appear as mostly cystic. At MR imaging, these tumors have intermediate to low signal intensity on T1-weighted images and intermediate to high signal intensity on T2-weighted images, depending on the amount of necrosis [1]. Mixed signal intensity and a fluid–debris level can be seen in hemorrhagic lesions [1]. The presence of extensive necrosis in a retroperitoneal mass, with contiguous involvement of a vessel, is highly suggestive of leiomyosarcoma [1]. Metastasis to the liver, lungs, or lymph nodes occurs late in the course of the disease [22, 32].

A330234_1_En_6_Fig6_HTML.gif


Fig. 6.6
Leiomyosarcoma in a 55-year-old woman. (a, b) Axial and coronal reformatted contrast-enhanced CT images incidentally discovered a solid-enhancing mass (arrows) containing internal cystic portion in left perirenal space. At that time, neurogenic tumor including neurilemmoma or paraganglioma was proposed as a radiological diagnosis. (c) After 7 months, the follow-up CT shows marked interval increase in size of the mass (arrow) with more heterogeneous enhancement and internal nonenhancing areas. Pathological examination after surgical resection confirmed leiomyosarcoma


6.3.3 Undifferentiated/Unclassified Sarcoma


This new category in the 2013 WHO classification, which may comprise up to 20 % of all pleomorphic soft tissue sarcomas [33], encompasses tumors in which all recognizable lines of differentiation have been excluded. Many of these tumors would have been called MFH in the past. Since MFH was first described as a distinct histologic type of soft tissue sarcomas in 1964, large series of cases were reported [34, 35]. However, MFH has been plagued by controversy in terms of both its histogenesis and its validity as a clinicopathologic entity. Many pathologists recognized that most so-called MFH located in the retroperitoneum were dedifferentiated liposarcomas and only a few cases may be classified as undifferentiated pleomorphic sarcoma [35]. The latest WHO classification no longer includes MFH as a distinct diagnostic category but rather as subtypes of an undifferentiated/unclassified sarcoma [16, 17]. These tumors are typically high grade, show a wide range of morphological features, and are often associated with a poor prognosis. These tumors can be subclassified according to predominant morphological patterns: round cell, spindle cell, pleomorphic, and epithelioid. Undifferentiated sarcoma, formerly known as MFH, is the third most common sarcoma in the retroperitoneal space and has been regarded as the most common soft tissue sarcoma in the whole body [1].

CT and MRI findings of the MFH were nonspecific and demonstrated a large, infiltrating, and heterogeneously enhancing soft tissue mass with areas of necrosis and hemorrhage and with possible invasion of adjacent organs (Fig. 6.7). Variable patterns of calcification can be seen (7–20 % of cases) in the peripheral portions of these tumors [1]. The presence of calcification may help to distinguish MFH from leiomyosarcoma [22]. The changes or controversial application in terminology and classification has represented a source of potential diagnostic confusion. Imaging interpretation should be updated with the latest pathological classification to reach the more specific diagnosis in predicting the histological type or grade of retroperitoneal tumors.

A330234_1_En_6_Fig7_HTML.gif


Fig. 6.7
Undifferentiated pleomorphic sarcoma in a 36-year-old woman. Contrast-enhanced CT ((a), axial; (b), coronal reformatted) shows a large well-circumscribed heterogeneous mass with internal necrotic areas at left retroperitoneal space. Direct tumor invasion of adjacent muscles, including the psoas and quadratus lumborum muscles, is shown, and the left kidney is displaced superiorly


6.3.4 Solitary Fibrous Tumor


Visceral pleura is the most common site of solitary fibrous tumor occurrence, but it can occur outside of the thoracic cavity. Retroperitoneal solitary fibrous tumors are rare, and their clinical manifestations depend on the location and size of the tumor. They grow slowly in an expansive way and displace adjacent structures by compression. For extrapleural solitary fibrous tumor, there is now complete omission of the prior synonym “hemangiopericytoma” according to the WHO classification of soft tissue tumors [16, 17].

The retroperitoneal solitary fibrous tumors were mostly lobulated and large, in round or oval shape (Fig. 6.8). Myxoid degeneration, hemorrhage, and necrosis may be visible, especially in huge masses; the tumor density or signal intensity became more heterogeneous on imaging. Rarely, some may have calcification. Most retroperitoneal solitary fibrous tumors show moderate or intense enhancement. Complete surgical resection is the standard therapy for solitary fibrous tumor, and pathologically negative resection margins are important to achieve a good prognosis.

A330234_1_En_6_Fig8_HTML.gif


Fig. 6.8
Solitary fibrous tumor in a 46-year-old man. (a) Transabdominal sonogram shows a large, lobulated mass (arrows) at left paravertebral region. (b) Contrast-enhanced axial CT at aortic bifurcation level demonstrates a large, lobulated mass (arrow) with intense enhancement at left anterior paravertebral region. Note direct tumor invasion into the adjacent left psoas muscle (small arrow)


6.3.5 Less Common Sarcomas


Rhabdomyosarcoma is the most common soft tissue sarcoma in children. The tumor is believed to arise from primitive muscle cells, but tumors can occur anywhere in the body. The retroperitoneum is involved in 7 % of cases diagnosed with rhabdomyosarcoma [36]. Cross-sectional imaging shows a mass with heterogeneous attenuation or signal intensity, focal calcifications, necrosis, and significant contrast enhancement.

Extraskeletal chondrosarcoma in retroperitoneum is very rare. The histologic types of lesions that account for extraskeletal chondrosarcoma are myxoid, mesenchymal, and very rarely low grade [37]. On CT and MR images, both myxoid and mesenchymal chondrosarcomas have features similar to those described for tumors of these histologic types located in bone [37]. Imaging of these lesions demonstrates a nonspecific soft tissue mass with areas of chondroid matrix mineralization, which may be seen as ring- and arc-like, stippled, and highly opaque calcifications (Fig. 6.9).

A330234_1_En_6_Fig9_HTML.gif


Fig. 6.9
Extraskeletal chondrosarcoma in retroperitoneum in a 51-year-old man. Contrast-enhanced CT scan of the abdomen shows a large soft tissue mass in the left retroperitoneum with invasion of the left psoas muscle. Note punctate or stippled calcifications (arrows) in the mass

Extraskeletal osteosarcoma is a rare malignant mesenchymal tumor characterized by the direct production of osteoid or bone by tumor cells without primary bone or periosteal involvement. The most common location of these tumors is the lower extremity, especially the thigh, followed by the upper extremity and the retroperitoneum [38]. Imaging shows a large soft tissue with areas of amorphous calcifications, necrosis, or old hemorrhage (Fig. 6.10). The imaging findings of extraskeletal osteosarcoma are nonspecific, with considerable overlap with other retroperitoneal masses. A wide range of differential diagnoses include several benign and malignant conditions that show mineralization, which include nontumoral benign conditions such as calcified hematoma or myositis ossificans and numerous benign and malignant retroperitoneal tumors.

A330234_1_En_6_Fig10_HTML.gif


Fig. 6.10
Extraskeletal osteosarcoma in a 64-year-old man. (a, b) Precontrast (a) and post-contrast (b) axial CT images show a well-defined soft tissue mass (arrow) at right retroperitoneum with abutting the right psoas muscle and ICV (arrowheads). Multiple amorphous calcifications are seen in the mass. (c) Axial T2-weighted MR images show heterogeneously hyperintense mass. (d) Coronal T1-weighted fat-suppressed MR images after administration of contrast material shows avid contrast enhancement with radiating pattern in the mass (arrow). The mass compresses infrarenal IVC without invasion. Right ureter was also compressed by the mass, with resultant obstructive hydronephrosis


6.4 Tumors of Neurogenic Origin


Neurogenic tumors constitute 10–20 % of primary retroperitoneal tumors in adult [1]. Compared with the tumors of mesodermal origin, they are generally benign, occur in a younger population, and have a better prognosis [1]. They are classified according to their cells of origin. They can originate from the nerve sheaths (schwannoma, neurofibroma, neurofibromatosis, malignant nerve sheath tumors), sympathetic nerves/ganglionic cells (ganglioneuroma, ganglioneuroblastoma, neuroblastoma), or chromaffin/paraganglionic cells (paraganglioma, pheochromocytoma). They are seen commonly along the sympathetic ganglia, which are located in the paraspinal region, and in the adrenal medulla or the organs of Zuckerkandl (para-aortic bodies) [1]. Less commonly, they occur in other sites, such as the urinary bladder, abdominal wall, bowel wall, or gallbladder [1, 39]. Neuroblastoma and ganglioneuroblastoma are malignancies of childhood and are very rare in adults [15].


6.4.1 Schwannoma


Schwannoma, also known as neurilemmoma, is the most common retroperitoneal neurogenic tumor and accounts for 4 % of all retroperitoneal tumors [5]. It is a solitary, slow-growing benign tumor that arises from peripheral nerve sheath and is usually located in the paravertebral or presacral region. Pathologically, it appears as a rounded or oval mass and is well encapsulated and extends along the course of a nerve, with the nerve of origin flattened against the periphery of the tumor [1]. Microscopically, it consists of two different components (Antoni A and Antoni B). Antoni A area is highly cellular, while Antoni B area is less cellular, with cystic and edematous area. At MR imaging, highly cellular areas are hypointense on T1- and T2-weighted images, while cystic and edematous areas appear hyperintense on T2-weighted images. Small schwannomas may be seen as round, well-circumscribed, and homogeneous masses, but large schwannomas have a heterogeneous appearance and are more likely to undergo degenerative changes (hemorrhage, necrosis or cystic changes, calcification, and hyalinization) (Fig. 6.11). Contrast enhancement is heterogeneous, with nonenhancing cystic components and enhancing solid components [1]. Malignant transformation rarely occurs [1, 40].

A330234_1_En_6_Fig11_HTML.gif


Fig. 6.11
Schwannoma: various patterns on axial contrast-enhanced CT images. (a) A small schwannoma in a 58-year-old woman is seen as a round, well-circumscribed solid mass (arrow) with slightly inhomogeneous enhancement at left retroperitoneal space of the periadrenal region. (b) Another case of small schwannoma in a 31-year-old woman is seen as an oval, well-circumscribed solid mass (arrow) with internal nonenhancing portion at retrocaval region. (c) A schwannoma in a 34-year-old woman is shown as a well-defined round mass (arrow) at left para-aortic region. The mass is seen to be predominantly cystic with septations. (d) Multiple schwannomas in a 6x-year-old woman are seen as bilateral retroperitoneal soft tissue masses (arrows) with prominent cystic degeneration. (e) A large schwannoma in a 54-year-old woman is seen as a heterogeneous presacral mass with internal cystic or necrotic changes (arrows) and neural foraminal widening and bone destruction (black arrows). (f) A benign schwannoma in a 48-year-old man demonstrates a multiloculated cystic mass with several septal calcifications (arrows) and extending into the left gluteal region through the greater sciatic foramen


6.4.2 Neurofibroma


Neurofibroma is a benign nerve sheath tumor that can occur as an isolated tumor (90 %) or as part of type 1 neurofibromatosis [1]. Approximately one-third of patients with a solitary neurofibroma are associated with type 1 neurofibromatosis, and almost every patient with multiple tumors or plexiform neurofibromas has type 1 neurofibromatosis [39]. Neurofibroma is more common in men, particularly in the 20–40-year age group [39]. Histopathologically, neurofibroma is an unencapsulated solid tumor that is composed of nerve sheath cells and collagen bundles with variable myxoid degeneration and causes expansion of the entire nerve, with nerve fibers traversing the tumor [1]. Cystic degeneration is rare. Plexiform neurofibroma is seen as a large extensive infiltrating mass and is commonly located in the lumbosacral plexus.

On imaging, neurofibroma is depicted as a well-defined, rounded mass with homogeneous contrast enhancement. Occasionally, it demonstrate target-like enhancement representing the center of nerve tissue and the periphery of myxoid degeneration. On T1-weighted images, the central portion of the tumor has higher signal intensity that is due to neural tissue, and on T2-weighted images, the periphery has higher signal intensity that is due to myxoid degeneration [1]. Tumors involving the neural foramina may show a “dumbbell” shape with widening of the bony foramina or vertebral body scalloping.


6.4.3 Malignant Peripheral Nerve Sheath Tumor


Malignant peripheral nerve sheath tumors include malignant schwannoma, neurogenic sarcoma, and neurofibrosarcoma [1]. Fifty percent of these tumors originate de novo, and the rest of them are derived from neurofibroma or ganglioneuroma or occur after exposure to radiation [39]. These are highly aggressive and infiltrative tumors, may present with distant metastases, and demonstrate high recurrence rate. They may be clinically asymptomatic, but progressive enlargement and pain can be suggestive of malignancy, especially when associated with type 1 neurofibromatosis. Although imaging is not reliable in differentiating between benign and malignant tumors, a heterogeneous nature, irregular or infiltrative borders with invasion into adjacent tissues and destruction of adjacent bones can be helpful for detecting malignancy (Fig. 6.12).
Jul 10, 2017 | Posted by in UROLOGY | Comments Off on Retroperitoneal Tumors

Full access? Get Clinical Tree

Get Clinical Tree app for offline access