Neoplasms of the Kidney



Fig. 35.1.
Renal cell carcinoma, clear cell type. Tumor is typically well demarcated from normal kidney by a fibrous pseudocapsule, and the cut surface is usually variegated. Viable tumor is golden yellow, in a background of cystic and myxoid degeneration, hemorrhage, and necrosis.




 




Microscopic



Typically composed of cells with abundant cytoplasm that appears optically clear (due to loss of intracellular glycogen and lipids during histologic processing) and with distinct cell membranes

 



Tumor architecture is diverse – solid, alveolar, and acinar patterns are often admixed

 



Tumor cells are set in a regular network of small delicate blood vessels (Fig. 35.2)

A145302_4_En_35_Fig2_HTML.jpg


Fig. 35.2.
Renal cell carcinoma, clear cell type. Classic clear cell carcinoma is composed of cells with distinct cell membranes and optically clear cytoplasm. Tumor cells are arranged in sheets, compact nests, alveolar, acinar, and microcystic or even macrocystic structures, separated by an abundance of thin-walled blood vessels.

 



Microcysts and macrocysts containing eosinophilic fluid may be present (Fig. 35.3)

A145302_4_En_35_Fig3_HTML.jpg


Fig. 35.3.
Renal cell carcinoma, clear cell type. Tubular structures vary in size. Microcysts contain eosinophilic fluid or extravasated red blood cells.

 



Many cytologic variations may be seen



  • Cells with granular eosinophilic cytoplasm


  • Cells with rhabdoid morphology


  • Interlacing or whorled bundles of spindle cells, sometimes with marked nuclear pleomorphism (sarcomatoid change)

 



ISUP nuclear grade is based on nucleolar prominence for grades 1–3 tumors; grade 4 tumors are defined by the presence of parameters listed below



  • Grade 1: nucleoli absent or inconspicuous at 400× (Fig. 35.4)

    A145302_4_En_35_Fig4_HTML.jpg


    Fig. 35.4.
    Clear cell renal cell carcinoma, ISUP grade 1. At 400× magnification, nucleoli are absent or inconspicuous.


  • Grade 2: nucleoli distinctly visible at 400× but inconspicuous or invisible at 100× (Fig. 35.5)

    A145302_4_En_35_Fig5_HTML.jpg


    Fig. 35.5.
    Clear cell renal cell carcinoma, ISUP grade 2. Nucleoli are inconspicuous or invisible at 100× but are distinctly visible at 400×, as shown here.


  • Grade 3: nucleoli are conspicuous at 100× (Fig. 35.6)

    A145302_4_En_35_Fig6_HTML.jpg


    Fig. 35.6.
    Clear cell renal cell carcinoma, ISUP grade 3. The large prominent nucleoli shown in this image (at 400×) were easily identified at 100× magnification, compatible with ISUP grade 3.


  • Grade 4: pronounced nuclear pleomorphism and/or tumor giant cells (Fig. 35.7) and/or rhabdoid morphology and/or sarcomatoid morphology (Fig. 35.8)

    A145302_4_En_35_Fig7_HTML.jpg


    Fig. 35.7
    Clear cell renal cell carcinoma, ISUP grade 4. Tumor cells with extreme pleomorphism are present.


    A145302_4_En_35_Fig8_HTML.jpg


    Fig. 35.8.
    Clear cell renal cell carcinoma , ISUP grade 4. Tumor cells show rhabdoid (left side of image) or sarcomatoid (right side of image) morphology.

 




Immunohistochemist ry (Table 35.1)



Incidence of immunoreactivity to antibodies against the following antibodies is as indicated:



  • PAX8 (95–100%)


  • Carbonic anhydrase IX (CA IX), 75–100%


  • Frequently positive: keratins AE1/AE3 and CAM5.2, vimentin, RCC marker, CD10, and EMA

 



Rarely expressed: CK7 and high molecular weight cytokeratins (14, 34βE12)

 



Table 35.1.
Immunohistochemical Findings in Renal Epithelial Neoplasms















































































































 
CD10

CA IX

RCC marker

Vimentin

EMA

CK7

CK20

HMWCK

CD117 (ckit)

AMACR

Clear cell renal cell carcinoma

+(81–100%)

+

+(85%)

+

+/− (20–90%)



−/+f

−/+f

−/+f

Papillary renal cell carcinoma

+(63–93%)

+/−f

+(93%)

+/−

+/− (30–45%)

+

+/−

−/+f

−/+f

+

Clear cell papillary renal cell carcinoma


+(cup-like)

+/−

+

+/−

+





Chromophobe renal cell carcinoma

−/+f


+(50%)


+

+



+


Oncocytoma

−/+f




+

Focal (5% or less)a

+/−


+

−/+f

Collecting duct carcinoma


+/−f


+

+/−

+/−


+



Urothelial carcinoma

−/+f

−/+



+

+

+

+

−/+f

−/+f


−/+f usually negative, occasionally focally positive; +/− frequently positive; AMACR alpha-methylacyl-CoA racemase (P504S); CK7 cytokeratin 7; CK20 cytokeratin 20; EMA epithelial membrane antigen; HMWCK high molecular weight cytokeratin 34βE12

a In contrast to diffuse and strong immunoreactivity for CK7 in chromophobe renal cell carcinoma, oncocytoma typically shows scattered focal positivity for CK7



Multilocular Cystic Renal Neoplasm of Low Malignant Potential




Clinical



Occurs in adults, with a mean age of 51 years (range, 20–76)

 



90% or more are discovered incidentally

 



Accounts for less than 1% of all renal tumors

 



Male to female ratio is about 2:1

 



No recorded instances of recurrence or metastasis after excision

 


Macroscopic



Well circumscribed, with fibrous capsule

 



Composed entirely of cysts containing clear, serous, gelatinous, or hemorrhagic fluid

 



Cysts are separated by irregular septa of variable thickness

 



Grossly evident solid nodules of tumor exclude the diagnosis

 


Microscopic



The cells lining the cysts form a single layer, although, occasionally, small papillae may extend into the cysts (Fig. 35.9)

A145302_4_En_35_Fig9_HTML.jpg


Fig. 35.9.
Multilocular cystic neoplasm of low malignant potential. Delicate septa between the cystic spaces are lined by clear cells (blue arrows) and flat cuboidal cells.

 



Tumor cells have plentiful clear cytoplasm and small nuclei that lack nucleoli (ISUP grade 1)

 



Septa consist of fibrous tissue, sometimes with calcific or osteoid deposits

 



Septa contain nonexpansile aggregates of clear cells similar to those that line the cysts (Fig. 35.10)

A145302_4_En_35_Fig10_HTML.jpg


Fig. 35.10.
Multilocular cystic neoplasm of low malignant potential. Aggregates of clear cells are present in the septa; the aggregates are noted only by microscopic examination; they are not “expansile”.

 



Sarcomatous change, vascular invasion, and necrosis exclude the diagnosis

 


Genetics



Probably genetically related to clear cell renal cell carcinoma

 



74% has chromosome 3p deletions, similar to those identified in 89% of clear cell renal cell carcinoma

 



VHL mutations have been found in 25% of cases

 


Papillary Renal Cell Carcinoma




Clinical



Male to female ratio is about 2:1; affects patients from pediatric age to great old age; mean patient age is 59–63 years

 



Accounts for up to 18.5% of all RCCs; it is the second most common type of renal cell carcinoma

 



Using the 2016 WHO criteria, papillary or tubulopapillary tumors greater than 1.5 cm in diameter are considered carcinoma

 



Unencapsulated tumors with papillary or tubular architecture of low nuclear grade (grades 1–2) and ≤1.5 cm in size are considered benign (papillary adenoma; see further discussion in the “Papillary Adenoma” section)

 



Hereditary papillary RCC is rare and is related to a germline mutation of the cmet gene

 



Cytogenetics



  • Trisomy or tetrasomy 7, trisomy 17, loss of chromosome Y most commonly


  • Trisomy of chromosomes 8, 12, 16, and 20 common


  • Wide variety of other chromosomal aberrations also reported

 



Prognosis



  • Overall, better than for carcinomas of clear cell, collecting duct, or HLRCC type


  • Prognosis for type 1 tumors is better than for type 2 tumors


  • Prognosis is worsened by the presence of sarcomatoid or rhabdoid morphology in the tumor

 


Macroscopic



Well-circumscribed cortical tumor, often with a thick fibrous pseudocapsule

 



Solid tumors often have a friable consistency (Fig. 35.11)

A145302_4_En_35_Fig11_HTML.jpg


Fig. 35.11.
Papillary renal cell carcinoma . A thick fibrous pseudocapsule is present in up to two-thirds of cases. Many such tumors are extensively hemorrhagic and friable in consistency. Infrequently the thick pseudocapsule encloses little more than old bloody necrotic material, and extensive tumor sampling is needed to identify viable tumor.

 



Hemorrhage, necrosis, and cystic degeneration are common

 



More often multiple than other types of RCC

 



May contain golden yellow areas reflecting macrophage content

 


Microscopic



Tumor is composed of papillary structures and tubules in varying proportions

 



Papillary cores are lined by a single cell layer that may show pseudostratification

 



Fibrovascular cores may be sclerotic, edematous, or expanded by foamy or hemosiderin-laden macrophages, psammoma bodies, or cholesterol clefts

 



Two morphologic variants



  • Type 1: characterized by a single layer of small dark cells with scant cytoplasm and low nuclear grade (Fig. 35.12)

    A145302_4_En_35_Fig12_HTML.jpg


    Fig. 35.12.
    Papillary renal cell carcinoma, type 1. Tumor is composed of papillary structures lined by small dark uniform cells with minimal cytoplasm and grade 1 nuclear characteristics.


  • Type 2: cells lining the papillae are large and pseudostratified, with more abundant pink cytoplasm, and of higher nuclear grade (Fig. 35.13)

    A145302_4_En_35_Fig13_HTML.jpg


    Fig. 35.13.
    Papillary renal cell carcinoma, type 2. Tumor is composed of papillary structures lined by pseudostratified cells with abundant pink cytoplasm and high-grade (grade 3) nuclear characteristics.

 



Tubules may be compacted to form “parallel arrays” (Fig. 35.14)

A145302_4_En_35_Fig14_HTML.jpg


Fig. 35.14.
Papillary renal cell carcinoma. The papillary structures are compressed and form “parallel arrays.” Arrow indicates stromal macrophages.

 



Papillary stalks often contain abundant foamy macrophages (Fig. 35.15)

A145302_4_En_35_Fig15_HTML.jpg


Fig. 35.15.
Papillary renal cell carcinoma . In this example, numerous foamy macrophages are present in the fibrovascular cores, a feature that is characteristic of this type of RCC.

 



Sarcomatoid change may be observed in up to 5% of cases

 



The ISUP grading system is applicable to papillary renal cell carcinoma

 


Immunohistochemistry



Tumor cells are frequently immunoreactive to antibodies against pancytokeratins, CK7, AMACR, vimentin, CD10, and RCC antigen

 


Hereditary Leiomyomatosis Renal Cell Carcinoma (HLRCC)-associated Renal Cell Carcinoma




Clinical



Rare and aggressive tumor

 



Arises in patients with an autosomal dominant tumor syndrome associated with germline mutations in the fumarate hydratase (FH) gene at chromosome 1q42

 



Syndrome is known as the hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome

 



Patients typically develop painful cutaneous leiomyomas, and females develop uterine leiomyomas, often at an unusually young age, and, rarely, uterine leiomyosarcomas

 



Diagnosis is confirmed by detection of germline mutations in the gene encoding fumarate hydratase (FH, 1q42.3-q43)

 



HLRCC syndrome has a lower penetrance of RCC; about 33% of patients with HLRCC syndrome develop renal cell carcinoma

 



Those who develop renal cell carcinoma typically present with advanced-stage disease, and the majority die of renal cancer

 



If the tumor is recognized, counseling and follow-up of family members can be initiated and may be greatly beneficial to them – identification and resection of even small tumors may be lifesaving

 


Macroscopic



With rare exceptions, tumors are unilateral and solitary; ranging in size from 2.3 cm to 20 cm

 



Tumors may be predominantly solid, predominantly cystic, or have a combination of solid–cystic components

 


Microscopic



Tumors typically exhibit papillary or tubulopapillary architecture

 



Morphology may closely mimic that of type 2 papillary renal cell carcinoma

 



Tumor cells have abundant eosinophilic cytoplasm and large nuclei with a very prominent inclusion-like orangiophilic or eosinophilic nucleoli, surrounded by a clear halo (Fig. 35.16)

A145302_4_En_35_Fig16_HTML.jpg


Fig. 35.16.
Hereditary leiomyomatosis renal cell carcinoma. Tumor morphology is similar to that of conventional type 2 papillary renal cell carcinoma, but many of the large nuclei exhibit very prominent inclusion-like orangiophilic or eosinophilic nucleoli, surrounded by a clear halo, a finding that should raise concern for this aggressive type of renal cancer.

 


Other studies



Immunohistochemistr y: loss of FH staining and overexpression of modified cysteine (2SC)

 



Somatic mutations in FH are rare in sporadic type 2 papillary renal cell carcinoma

 


Chromophobe Renal Cell Carcinoma




Clinical



Accounts for about 5–7% of all resected renal cell carcinomas

 



Believed to originate from intercalated cells of collecting duct epithelium

 



Males and females are almost equally affected; mean patient age is about 55 years

 



Cytogenetics: Typically, extensive loss of chromosomes (−1, −2, −6, −10, −13, −17, −21) with hypodiploid DNA index

 



Commonly of low stage when detected

 



Outcome predicated by tumor stage and the presence or absence of sarcomatoid transformation, tumor necrosis, and small vessel invasion

 



Overall prognosis is relatively favorable: reported 5 year survival ranges from 78% to 100%

 


Macroscopic



Typically solid, light tan or mahogany brown, and well circumscribed (Fig. 35.17)

A145302_4_En_35_Fig17_HTML.jpg


Fig. 35.17.
Chromophobe renal cell carcinoma. A solitary well-circumscribed tumor with a solid, homogeneous, and light brown cut surface which is shown here. Less often the tumor is gray tan, light pink, or yellow white.

 



Necrosis, hemorrhage, and scarring are minimal or absent

 


Microscopic



Growth pattern is generally solid but sometimes limited tubule formation is evident

 



Sheets of cells are intersected by fibrovascular septa, which may be thin and delicate but are often broad and hyalinized

 



Tumors are composed of varying proportions of two cell types



  • “Classic cell type” (Fig. 35.18):

    A145302_4_En_35_Fig18_HTML.jpg


    Fig. 35.18.
    Chromophobe renal cell carcinoma, “classic cell type”: large polygonal cells with thick distinct “plant-like” cell membranes and abundant pale or translucent flocculent pink cytoplasm.




    • Large polygonal cells, with thick distinct “plant-like” cell membranes


    • Tumor cells have abundant pale or translucent flocculent pink cytoplasm


    • The largest tumor cells tend to aggregate adjacent to fibrovascular septa




  • “Eosinophilic cell type” (Fig. 35.19):

    A145302_4_En_35_Fig19_HTML.jpg


    Fig. 35.19.
    Chromophobe renal cell carcinoma, “ eosinophilic cell type.” Cytoplasm is less abundant and is darkly eosinophilic. Blue arrow indicates a round regular nucleus with characteristic perinuclear halo; black arrow indicates a mitotic figure; green arrow indicates an irregular “raisinoid” nucleus.




    • Smaller cells, arranged in small nests, or intermingled with “classic” cells


    • Cytoplasm is less abundant and is granular and more darkly eosinophilic


    • Distinctive perinuclear halos are often present due to the accumulation of cytoplasmic organelles around nuclei

 



In either cell type, nuclei may be round and uniform or irregular and “raisinoid”

 



ISUP grading is not valid for this type of renal cell carcinoma

 



Sarcomatoid transformation is observed in 2–8% cases; the presence of this finding markedly worsens the prognosis

 


Histochemical and Immunohistochemical Studies



Hale colloidal iron stain for acid mucopolysaccharides variable but strongly and diffusely positive in many cases; technically a difficult stain to perform, interpretation may be difficult as well

 



Typically positive immunostaining for CK7, CD117, parvalbumin, and kidney-specific cadherin

 



Usually negative immunostaining for vimentin

 


Ultrastructure



Numerous cytoplasmic microvesicles (150–300 nm) of unknown origin

 


Collecting Duct Carcinoma




Clinical



Accounts for less than 1–2% of renal cancers

 



Male to female ratio is about 2:1; mean age is about 55 years (range, 13–85 years)

 



Nearly 33% of patients have metastases at the time of diagnosis and more than 70% are at pathologic stage pT3 or higher when diagnosed

 



Cytogenetics: loss of heterozygosity on multiple chromosome arms (1q, 3p, 6p, 8p, 13q, 21q) and losses of entire chromosomes are but a few of a wide variety of reported abnormalities

 



Prognosis: Poor; about 67% of patients die within 2 years after diagnosis

 


Macroscopic



Tumors are typically gray white and range in size from 2.5 cm to 15 cm, with an average of 5 cm

 



When small, they are centered in the renal medulla

 



Tumors exhibit irregular and grossly infiltrative borders

 


Microscopic



Composed of tubular or tubulopapillary structures

 



Lined by cytologic malignant cells, some may show a hobnail appearance

 



Irregular angulated glands infiltrate renal parenchyma

 



Infiltration is accompanied by prominent stromal desmoplasia and frequently by a pronounced chronic inflammatory response at the interface between advancing tumor and normal adjacent renal parenchyma (Figs. 35.20 and 35.21)

A145302_4_En_35_Fig20_HTML.jpg


Fig. 35.20.
Collecting duct carcinoma . This low power view illustrates the tubular architecture of the tumor, stromal desmoplasia, and the “pushing” border, with pronounced inflammation (blue arrows) at the interface between tumor and normal kidney.


A145302_4_En_35_Fig21_HTML.jpg


Fig. 35.21.
Collecting duct carcinoma . Tumor is composed of irregular tubules in a desmoplastic stroma (blue arrows), with considerable peritumoral chronic inflammatory cell infiltrate (green arrows).

 



Intraluminal and intracytoplasmic mucin may be seen

 



Dysplasia may be noted in the epithelium of adjacent collecting ducts (Fig. 35.22)

A145302_4_En_35_Fig22_HTML.jpg


Fig. 35.22.
Collecting duct carcinoma , with high-grade nuclear dysplasia in some native renal tubules (blue arrows); normal tubules are indicated by green arrows.

 



Sarcomatoid change is common, occurring in up to 30% of cases

 



Difficult to separate from urothelial carcinoma, adenocarcinoma of renal pelvis, metastatic adenocarcinoma, renal medullary carcinoma, and type 2 papillary renal cell carcinoma

 


Immunohistochemistry



Typically expresses high molecular weight keratins: CK7, CK19, and 34βE12

 



Recommended panel to differentiate from mimics: PAX2, PAX8, INI1, OCT3/4, and p63

 


Renal Medullary Carcinoma




Clinical



Most often occurs in young persons with sickle cell trait, sickle cell disease, or hemoglobin sickle cell disease

 



Male to female ratio is about 2:1

 



The great majority of patients (90%) are African-American, Hispanic, or Brazilian; only 5% is Caucasian

 



Age range is 5–69 years; most are in second and third decades of life

 



Weight loss, hematuria, flank or abdominal pain, or palpable mass are common symptoms and signs

 



Often, the presenting symptoms are related to metastases

 



Tumor is believed to originate in collecting duct epithelium

 



Genetics: increased HIF1A expression, deregulated DNA remodeling and repair, LOH or deletions at SMARCB1, and some similarities to urothelial carcinoma

 



Prognosis is poor: most patients die within 1 year of diagnosis, and mean survival is only about 15 weeks

 


Macroscopic



Gray white, grossly infiltrative, and typically centered in the renal medulla, 4–12 cm (mean, 7 cm), with a predilection for right kidney

 


Microscopic



A variety of architectural patterns may be present



  • Reticular or yolk-sac appearance


  • Adenoid cystic appearance


  • Solid sheets, nests, or tubules

 



Tumor cells may show rhabdoid morphology

 



A strikingly prominent infiltrate of neutrophils may be evident within the tumor, and mucin production by tumor cells is often observed (Fig. 35.23)

A145302_4_En_35_Fig23_HTML.jpg


Fig. 35.23.
Renal medullary carcinoma . This type of RCC commonly demonstrates several different morphologic patterns. The malignant tubules in this example are similar to those seen in collecting duct carcinoma.

 



Sickled erythrocytes are evident in the great majority of cases (Fig. 35.24)

A145302_4_En_35_Fig24_HTML.jpg


Fig. 35.24.
Renal medullary carcinoma . Arrows indicate sickled red cells in a background of poorly differentiated renal carcinoma.

 


Immunohistochemistry



Tumor cells usually show positive immunostaining for PAX8, CK7, polyclonal CEA, keratin CAM5.2, and frequently OCT3/4

 



INI1 immunostain is negative in tumor cells due to deletions at SMARCB1 locus

 


MIT Family Translocation Renal Cell Carcinoma






TFE3 and TFEB are members of the MIT subfamily of transcription factors

 



Gene fusions involving TFE3 are associated with Xp11 translocation carcinomas of the kidney

 



A MALAT1TFEB gene fusion is associated with t(6;11) translocation carcinomas of the kidney

 


Xp11 Translocation Renal Cell Carcinoma



Accounts for about 40% of pediatric renal cell carcinomas and up to 4% of adult renal carcinomas

 



Tumors are defined by a variety of chromosome translocations , all of which involve a breakpoint at Xp11.2 and all of which result in the fusion of any one of multiple translocation partners with the TFE3 transcription factor gene at this locus



  • t(X;1)(p11.2;q21) translocation fuses the PRCC and TFE3 genes


  • t(X;1)(p11.2;p34) fuses the PSF and TFE3 genes


  • inv(X)(p11;q12) fuses the NonO (p54 nrb ) and TFE3 genes


  • t(X;17)(p11.2;q23) fuses the CLTC and TFE3 genes

 



A balanced t(X;17)(p11.2;q25) in one variant of translocation carcinoma; it shares the same ASPSCR1TFE3 gene fusion as alveolar soft part sarcoma – der(17)t(X;17)(p11.2;q25), which fuses the TFE3 gene to a novel gene, ASPL, on 17q25

 


Clinical



Majority of patients are less than 50 years old, but patients up to 79 years old have been reported

 



Equally frequent in males and females

 



Some patients have a history of prior chemotherapy treatment

 



Half of patients present with abdominal or flank pain or hematuria; some tumors are detected as a palpable mass, and some are found incidentally

 



Prognosis: Similar to that of patients with clear cell RCC but worse than for those with papillary RCC

 



Outcome is worse in older patients and in those with metastatic disease at presentation

 


Morphology



Circumscribed 3–14 cm and may or may not be encapsulated

 



Typically, papillary and nested architecture; some have abundant psammoma bodies (Fig. 35.25)

A145302_4_En_35_Fig25_HTML.jpg


Fig. 35.25.
Xp11 translocation renal cell carcinoma (ASPSCR1TFE3 type). Tumor shows nested to pseudopapillary architecture and numerous psammomatous calcifications . Tumor cells possess voluminous clear cytoplasm. When examined at higher power, they have vesicular nuclei with prominent nucleoli.

 



Tumor cells are epithelioid, with sharply defined cell borders and with abundant cytoplasm that varies from clear and finely granular to distinctly eosinophilic

 



Areas of necrosis are seen in the majority of cases, but mitotic activity is very limited

 



In rare cases, melanin pigment may be present

 


Immunohistochemistry and FISH



Cytokeratins and EMA are underexpressed

 



PAX8 consistently positive

 



Cathepsin K positive in 60%; melanocytic markers positive rarely

 



Strong nuclear immunostaining for TFE3 is highly sensitive and specific but technically difficult in formalin-fixed paraffin-embedded (FFPE) tissues

 



TFE3 break-apart FISH assay is less prone to fixation problems

 


t(6;11) renal cell carcinoma



Characterized by a t(6,11)(p21;q12) translocation that results in fusion of the 5′ portion of the alpha gene with the transcription factor gene TFEB at 6p21

 


Clinical



Less common than Xp11 translocation RCC; fewer than 50 reported to date

 



Mean and median age, 31 years (range, 9–60 years)

 



About half of patients present with abdominal pain or hematuria; the rest are asymptomatic

 



Typically low stage at presentation (T1 or T2)

 



Majority exhibit indolent biologic behavior

 



Less than 10% of reported cases have had adverse outcome (metastasis and death from cancer)

 


Morphology



Typically solid, focal cyst formation, pseudoencapsulated, homogeneous tan–yellow–brown color, without evidence of necrosis

 



Size ranges from 2 cm to 12 cm in greatest dimension (mean diameter, 7 cm)

 



Entrapped single native renal tubules characteristically evident at the periphery of the tumor

 



Solid, nested pattern of growth, biphasic cell population

 



Predominant cell type is epithelioid, with well-defined borders, abundantly clear, finely granular, or eosinophilic cytoplasm and rounded or vesicular nuclei that have prominent nucleoli evident at low magnification

 



In all cases, a second cell population comprising 5–30% of the tumor and consisting of smaller cells with denser chromatin, usually clustered around nodules of hyaline basement membrane material (Fig. 35.26)

A145302_4_En_35_Fig26_HTML.jpg


Fig. 35.26.
t(6;11) translocation renal cell carcinoma.

 



Mitotic figures absent or rare

 


Immunohistochemistry and FISH



Cytokeratins and EMA are underexpressed

 



PAX8 consistently positive

 



Consistently expresses cathepsin K and melanocytic markers Melan A and HMB45

 



Nuclear immunoreactivity for TFEB is highly specific

 



TFEB break-apart FISH is the preferred method of confirming the diagnosis in FFPE tissue

 


Renal Cell Carcinoma Associated with Neuroblastoma






Very rare; fewer than 10 cases

 



In all instances, renal cell carcinoma arose 3 to 35 years after the child was treated for neuroblastoma at age 2 years or less

 



Although most were treated with chemotherapy and/or radiation therapy, at least two patients never received either form of treatment, and the etiology of the renal cancer may be on the basis of genetic susceptibility, rather than treatment induced

 



A few cases of renal cancer after neuroblastoma treatment have been shown to have Xp11 translocation, which has a known association with prior chemotherapy

 



Tumor size: 1–8 cm; rarely bilateral

 



Stage at time of diagnosis varies from localized to distantly metastatic

 



Tumors are heterogeneous, but generally cells are arranged in solid sheets or nests and in papillae, sometimes intermingled with clusters of macrophages and sometimes with psammoma bodies

 



Tumor cells have sharply defined cell membranes and abundant eosinophilic granular cytoplasm, giving it an “oncocytoid” appearance. In addition, aggregates of cells with abundant reticular or vacuolated cytoplasm are present, with some morphologic resemblance to chromophobe cells (Fig. 35.27)

A145302_4_En_35_Fig27_HTML.jpg


Fig. 35.27.
Renal cell carcinoma associated with neuroblastoma.

 



Nuclear morphology and grade are variable among individual tumors

 



Immunoprofile



  • Positive for EMA, vimentin, and cytokeratins 8 and 18


  • Negative for cytokeratins 7, 14, 19, and 20


  • Negative for HMB45, TFE3, TFEB, and S100 protein

 



Genetics: Clonal abnormalities, none of which are consistent or recurrent

 



Prognosis: Cases lacking high-grade cytologic atypia have shown indolent behavior. High stage portends a less favorable outcome

 


Mucinous Tubular and Spindle Cell Carcinoma




Clinical



Rare tumor, less than 1% of renal neoplasms

 



Originally described as a subset of “low-grade collecting duct carcinoma”

 



Generally asymptomatic, and most are found incidentally

 



Female predilection: M:F ratio is about 1:3; mean patient age is 58 years, with a range of 13–82 years

 



Cytogenetics: Consistent losses involving chromosomes 1, 4, 6, 8, 9, 13, 14, 15, and 22

 



Biologic behavior is indolent in most cases

 



Although rare, recurrence, lymph node metastasis, distant metastasis, and death due to cancer have been reported, in cases with typical bland morphology and in cases with high-grade cytologic features

 


Macroscopic



Well-circumscribed, gray or light tan, and glistening cut surface (Fig. 35.28)

A145302_4_En_35_Fig28_HTML.gif


Fig. 35.28.
Mucinous tubular and spindle cell carcinoma. Tumor is a sharply circumscribed gray-white neoplasm with central degeneration and a slightly bulging glistening cut surface.

 


Microscopic



Tightly packed elongated tubules, arranged in parallel with some branching (Fig. 35.29)

A145302_4_En_35_Fig29_HTML.jpg


Fig. 35.29.
Mucinous tubular and spindle cell carcinoma. Elongated tubules in a bubbly myxoid stroma consisting of abundant extracellular mucin.

 



Abundant pale eosinophilic or basophilic extracellular mucin (Fig. 35.30)

A145302_4_En_35_Fig30_HTML.jpg


Fig. 35.30.
Mucinous tubular and spindle cell carcinoma. Higher power view of the tumor shown in Fig. 35.42, illustrating the extremely bland nuclear characteristics of the tumor, possibly accounting for its very indolent behavior.

 



Collapsed tubules may impart a spindled appearance

 



Tumor cells are usually cytologically low grade, round or oval, although rare cases with high-grade/sarcomatoid features have been reported

 


Immunohistochemistry



Most express CK7 and AMACR

 


Tubulocystic Renal Cell Carcinoma




Clinical



A rare tumor, less than 1% of renal carcinomas

 



Originally described as a subset of “low-grade collecting duct carcinoma”; assessment of cell of origin presently under investigation

 



Two-thirds of patients are asymptomatic, and the tumor is found incidentally

 



Male to female ratio is about 6:1; mean patient age is 58 years with a range of 30–94 years

 



Cytogenetics: Gains of 7 and 17 and loss of Y

 



Tumor is relatively indolent; survival rate exceeds 90%; one recurrence and four instances of metastasis in 70 reported cases

 



Tumors are generally low stage when initially detected

 


Macroscopic



Well circumscribed, with a mean size of 4.2 cm (range, 0.7–17 cm)

 



Spongy “bubble wrap-like” cut surface; grossly composed of innumerable cysts of variable size

 


Microscopic



Cystically dilated tubules of variable size, separated by thin “spidery” septa (Fig. 35.31) and lined by a single layer of flat, cuboidal, or hobnail cells (Fig. 35.32)
Sep 21, 2016 | Posted by in PATHOLOGY & LABORATORY MEDICINE | Comments Off on Neoplasms of the Kidney

Full access? Get Clinical Tree

Get Clinical Tree app for offline access