The Cytogenetics of Solid Tumors


Tumor

Chromosomal aberration(s)

Gene(s) involved

Clinical significance

References

Glial

Pilocytic astrocytoma

Gain of 5, 7/7q common, followed by 6, 8, 11, 12, 17, 19, 22

KIAA1549-BRAF from BRAF tandem duplication

Constitutive kinase activity

[35]

Diffuse grade II/anaplastic astrocytoma grade III

Loss of 10q, 13q, 17p, 19q

IDH1, IDH2, RB1 and TP53 mutation

TP53 mutation correlates with IDH mutations

[6]

Glioblastoma

Loss of 9p, 10q; EGFR amplification, MDM2 amplification

CDKN2A/B, PTEN, EGFR, MDM2

Short survival, aggressive course

[810]

Oligodendroglial

Oligodendroglioma (OD)

der(1;19)(q10;p10), loss of 1p, 19q

1p36, 19q13.3, IDH mutation

Longer survival, sensitive to therapy

[1115]

Anaplastic OD

der(1;19)(q10;p10), loss of 1p, 19q, 9p

1p36, 19q13.3, CDKN2A/B, IDH

Longer survival, sensitive to therapy

[1115]

Oligoastrocytic

Oligoastrocytoma (OA)

der(1;19)(q10;p10), TP53 mutation

1p36, 19q13.3, TP53, IDH mutation

Outcome is better with der(1;19)

[1, 16]

Anaplastic OA

der(1;19)(q10;p10), loss of 9p, 10q

1p36, 19q13.3, TP53, CDKN2A/B

Overall survival is better with der(1;19)

[20, 21]

Ependymoma

Spinal

Loss of 22q
 
More common in adults

[22]

Posterior fossa

Gain of 1q, balanced karyotypes
 
Young age, unfavorable prognosis

[23, 24]

Posterior fossa

Loss of 6q, 22q, gain of 9q, 15q, 18q
 
Favorable prognosis

[23, 24]

Intracranial

Gain of 1q, 7, loss of 6q, 9p

EGFR, CDKN2A

Gain of 1q, CDKN2A homozygous deletion is unfavorable

[25]

Choroid plexus

Papilloma (CPP)

Normal karyotype
 
Distinguish from atypical CPP

[2628]

Atypical papilloma

Hyperdiploid with gain of 7, 12, 20

Chromosome enumeration

Distinguish from CPC

[2628]

Carcinoma (CPC)

Hyperhaploid with gain of 1, 12, 20, 4, 7

Chromosome enumeration

Distinguish from papilloma

[2628]

Embryonal

Medulloblastoma, pediatric

Gain of 6q, MYC or MYCN amplification

MYC, MYCN, ERBB2

Very high risk, correlates with large cell/anaplastic histology

[2933]

i(17q), gain of 17q
 
Intermediate risk

[2933]

Loss of 6 or 6q
 
Excellent prognosis, desmoplastic histology

[2933]

Medulloblastoma, adult

Loss of 10q with gain of 17q, CDK6 amplification

CDK6 (7q21.2)

High risk, poor OS

[30]

Either loss of 10q or gain of 17q
 
Intermediate risk

[30]

Neither loss of 10q nor gain of 17q
 
Good prognosis

[30]

PNET, supratentorial

Gain of 1q, loss of 14, 16, 19, rare EGFR amplification

EGFR

Lacks i(17q), poor prognosis

[31, 35]

Atypical teratoid/rhabdoid tumor

Loss of 22, del(22q)

SMARCB1-INI1

Distinguishes AT/RT from MB, PNET, CPC

[3639]

Meningioma

Loss of 22, del(22q)

22q, NF2

Primary abnormality

[43, 44]

Atypical/anaplastic

loss of 1p, 14/14q, 18, 10, 6q
 
Increased risk of recurrence

[43, 44]


CPC choroid plexus carcinoma, CPP choroid plexus papilloma




Gliomas


Gliomas, the most common primary central nervous system (CNS) tumors, include astrocytomas, oligodendrogliomas, and ependymomas. Histopathologic features used for pathological classification and grading of the tumor correlate with prognosis and guide therapy. The annual incidence of primary brain tumors is ∼8–12 in 100,000 and for intraspinal tumors it is 1–2 in 100,000. CNS tumors are the most common (∼20%) cancers of childhood [1].


Astrocytomas


Astrocytomas have a spectrum of histologic features that correlate with the clinical course [2]. Grade I, pilocytic astrocytoma (PA), is a slow-growing, noninfiltrating tumor with a relatively benign course. Most PAs have a normal diploid karyotype; ∼30% show gain of one or more chromosomes. Approximately half of children younger than 15 years old show a single extra chromosome, while those greater than 15 years of age show gain of multiple chromosomes. Gains of chromosomes 5 and 7 are the most frequent, followed by chromosomes 6, 8, 11, 12, 17, 19, and 22. See Fig. 16.1. Tandem duplication of BRAF at7q34 is found in >50% of pediatric PA and in up to 66% of all PA [3, 4]. Tandem duplication produces a novel fusion gene, KIAA1549-BRAF, which has constitutive kinase activity [4, 5].

A145460_3_En_16_Fig1_HTML.jpg


Fig. 16.1
Pilocytic astrocytoma, grade I, from the posterior fossa of a 12-year-old boy: 47,XY,+5,der(14;21)(q10;q10)

Grade II diffuse astrocytoma and grade III anaplastic astrocytoma show increasing cellularity and nuclear pleomorphism. Gliomas of World Health Organization (WHO) grade II or III are invasive, progress to higher-grade lesions, and have a poor outcome. As the tumor progresses, acquired genetic abnormalities include loss of 10q, 13q, 17p, and 19q, and TP53 and RB1 mutations. The majority of WHO grade II and III gliomas, including astrocytomas and oligodendrogliomas, have IDH1 (2q34) or IDH2 (15q26.1) mutations. Most also have TP53 mutation. Patients with mutations of IDH genes have longer median overall survival, 65 versus 20 months [6].

Grade IV, glioblastoma (GB), is the most malignant astrocytoma. In addition to increases in cellularity, nuclear atypia, and pleomorphism, mitotic activity, vascular proliferation, and necrosis are characteristic. Genetic changes in primary glioblastomas, which account for >90% of GBs, include loss of 9p (CDKN2A deletion), loss of 10q (PTEN deletion), PTEN mutation, frequent EGFR amplification, and infrequent MDM2 amplification. The secondary glioblastomas (5–10%) that result from progression of grade II–III tumors have mutations of IDH1 or IDH2 and TP53. Tumors with wild-type IDH1 and IDH2 have fewer TP53 mutations and frequent alterations of PTEN, EGFR, CKDN2A, or CDKN2B [1, 68]. Median overall survival in patients with IDH mutations is twice that of patients with wild-type IDH, 31 versus 15 months [6]. Double minutes (dmin) with EGFR amplification have been demonstrated in up to 50% of GBs [9]. Current clinical trials are evaluating several targeted molecular therapies [10].


Oligodendroglial Tumors


Oligodendroglioma (OD), a diffusely infiltrating well-­differentiated (grade II) to anaplastic (grade III) glioma, is a relatively rare primary brain tumor, comprising 2–5% of all primary brain tumors. ODs characteristically show an unbalanced der(1;19)(q10;p10) with loss of 1p and 19q (Fig. 16.2). These aberrations may be detected by conventional chromosome analysis, fluorescence in situ hybridization (FISH), loss of heterozygosity (LOH) studies, or microarray comparative genomic hybridization (aCGH) [11, 12]. 1p/19q codeletion is associated with a longer median survival (∼10 years vs. 2 years) in patients with OD and anaplastic OD [13]. Anaplastic oligodendrogliomas show loss of 9p (CDKN2A/B) in additional to 1p/19q loss [14]. Recent studies have shown that IDH1 mutations are strongly associated with 1p/19q codeletion (∼85%) and that IDH1 mutation and 1p/19q codeletions are independent prognostic factors [6, 15].

A145460_3_En_16_Fig2_HTML.jpg


Fig. 16.2
Oligodendroglioma, grades II–III, from the brainstem of an 8-year-old girl: 57,XX,+der(1;19)(q10;p10)x2,+2,+3,+4,+8,+11,+16,+20,+20,+22. Arrows indicate the der(1;19); (see text for details)


Oligoastrocytic Tumors


Oligoastrocytomas (OA) are composed of a mixture of distinct cell types found in oligodendroglioma and astrocytoma. Genetic analysis reveals 1p/19q codeletion in ∼30–50%, TP53 mutations in ∼30%, and IDH mutations in 100% of OA. Progression-free survival (PFS) was 60 versus 30 months in patients with and without 1p/19q codeletion, respectively [1, 16]. Few studies have attempted to evaluate the genetics of the distinct cellular components. Rare tumors have been shown to have different genetic aberrations in the two histologic elements [1719]. Anaplastic OA (grade III) acquire additional genetic alterations—e.g., 9p (CDKN2A/B) loss, which is also implicated in the progression of astrocytoma and OD. Recent studies of anaplastic OD and OA confirm that presence of 1p/19q codeletion correlates with significantly longer survival than presence of genetic abnormalities other than 1p/19q codeletion [20, 21].


Ependymomas


Ependymomas are slow-growing neuroepithelial neoplasms that arise from the wall of the intracranial ventricles and the central canal of the spinal cord. Little is known about the genetics of myxopapillary ependymoma, a grade I tumor that arises predominately in the terminal spinal cord of young adults. The grade II ependymoma may arise at any site in the ventricular system and spinal canal with the posterior fossa the most common site in children. The most frequent cytogenetic aberrations include losses of 22q and 6q and gains of 1q and 9q (Fig. 16.3).

A145460_3_En_16_Fig3_HTML.jpg


Fig.16.3
Ependymoma, grade II, from the posterior fossa of a 2-year-old girl: 46,XX,+14,−22

Alterations of 22q are more frequently identified in spinal tumors. NF2 may be a candidate tumor suppressor gene in spinal ependymoma. However, few mutations in candidate genes, including NF2, TP53, PTEN, MEN2, and CDKN2A, have been identified in pediatric intracranial tumors [1, 3, 22].

Recent studies have identified two clinically and genetically different posterior fossa subgroups [23, 24]. Group A patients are younger (median age 4 years), more often male, more often have higher grade tumors (grade III), have more metastases at recurrence, and have a diminished prognosis compared with group B patients. Group A tumors have predominately balanced karyotypes; some show gain of 1q. Group B tumors frequently show loss of 6q and 22q and gain of 9q, 15q, and 18q. Intracranial tumors are more common in children than adults, with spinal tumors more common in adults. Tumors with gain of 1q correlate with higher grade, tumor recurrence, and worse prognosis [25].


Choroid Plexus Tumors


Choroid plexus (CP) tumors arise from the choroid plexus in the ventricles of the brain. Choroid plexus papilloma (CPP) is a benign (grade I) neoplasm with very low mitotic activity that closely resembles normal choroid plexus. Atypical CPP (grade II) is a CPP with increased mitotic activity that may have increased cellularity and nuclear pleomorphism. Grade III choroid plexus carcinoma (CPC) has frequent mitoses, increased nuclear pleomorphism, cellular density, necrotic areas, and sheets of tumor cells. The cytogenetics of these lesions helps to distinguish them from each other and from entities with which CP tumors may be confused. The CPP is karyotypically normal, the atypical CPP is hyperdiploid (Fig. 16.4), while CPC is hyperhaploid (Fig. 16.5). The most common recurrent chromosomal gains in atypical CPP are of chromosomes 7, 12, and 20, followed by gains of whole chromosomes 8, 9, 11, 15, 17, 18, and 19. Hyperhaploid CPCs characteristically have between 32 and 35 chromosomes; the most common gains (relative to a haploid background) in decreasing frequency order are of chromosomes 1, 12, 20, 4, and 7 [2628].

A145460_3_En_16_Fig4_HTML.jpg


Fig. 16.4
Choroid plexus papilloma, grade II, from the lateral ventricle of an 8-month-old boy: 55,XYY,+1,+5,+7,+8,+8,+12,+19,+20. The constitutional karyotype is 47,XYY


A145460_3_En_16_Fig5_HTML.jpg


Fig. 16.5
Choroid plexus carcinoma, grade III, from the lateral ventricle of a 4-month-old boy: 30<1n>,XY,+1,+2,+4,+12,+20,+21


Embryonal CNS Tumors



Medulloblastoma and Supratentorial Primitive Neuroectodermal Tumor


Medulloblastoma (MB) is a primitive small round cell tumor, which may show glial or neuronal differentiation. MB, also referred to as an infratentorial primitive neuroectodermal tumor (iPNET), is located in the cerebellum. MB is the most common malignant brain tumor in children, accounting for ∼20% of all pediatric brain tumors. MB is rare in adults, comprising ∼1% of primary adult intracranial malignant tumors. Supratentorial primitive neuroectodermal tumors (sPNETs) are histologically very similar to MBs, but are located in the cerebrum, and are clinically more aggressive.

Cytogenetically, i(17q) is the most common chromosome abnormality in pediatric MBs, present in ∼35% of cases, but it is not found in pediatric sPNET. Recent studies identified characteristic and prognostic cytogenetic subgroups [2931]. Highest risk pediatric MBs are characterized by gain of 6q, MYC amplification, MYCN amplification, and ERBB2 amplification; intermediate risk MB by 17q gain; and low risk MB by loss of 6q [29, 32]. High-risk genetic abnormalities strongly correlate with the large cell/anaplastic histology found in ∼7% of MBs [29, 33]. MYC/MYCN amplification is rare (∼4–10% of MBs). Patients with low-risk genetic abnormalities, loss of chromosome 6 or 6q, and nodular desmoplastic histology have an excellent 5-year overall survival (OS) of ∼90%. Most (∼90%) MBs have classic histology with a 60% OS. Gain of chromosome 7, common in MB, is not associated with OS [34].

Cytogenetic abnormalities associated with poor OS in adult MBs include high-level amplification of the oncogene CDK6 at 7q21.3 (more frequent than MYC), chromosome 17 aberrations including i(17q), and loss of 10q [30]. Oncogene amplification frequently associates with aberration of chromosome 17. Independent significant predictors for poor prognosis are loss of 10q and gain of 17q. Combined 10q loss and 17q gain show the poorest OS of ∼16%; either 10q loss or 17q gain is associated with a 44% OS, and absence of both 10q loss and 17q gain is associated with the best OS (92%).

Supratentorial PNETs are less well characterized due to a smaller number of sPNETs as compared with MBs. The most frequent cytogenetic aberrations so far associated with sPNET are gain of 1q and losses of 14q, 16p, and 19p/q. Gain of 17q is not found in sPNET. Rare amplifications of 1q, 4q12-q13, 8q22-q24, 19q12-q13, and EGFR are reported [31, 35].


Atypical Teratoid/Rhabdoid Tumor


Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant tumor in young (<5 years of age) children that may arise infratentorially (posterior fossa) or supratentorially. AT/RTs account for ∼10% of CNS tumors in infants. The tumor is defined by the rhabdoid cell, which resembles a rhabdomyosarcoma cell. Pathologically, AT/RTs may be difficult to recognize by histology alone due to variable components of primitive neuroectodermal, mesenchymal, and epithelial features. AT/RT may be distinguished from other poorly differentiated and anaplastic CNS tumors (e.g., sPNET, MB, or ependymoma) by a characteristic loss of chromosome 22 or 22q deletion [36]. The region of loss includes the SMARCB1/INI1 gene at 22q11.23. The overall prognosis for patients with AT/RT is poor, particularly in patients diagnosed at <3 years of age. AT/RT may be sporadic or part of the rhabdoid tumor predisposition syndrome [36, 37]. Germline deletion or mutation of SMARCB1 has been identified in up to 35% of patients with rhabdoid tumors [38]. Investigation of familial cases has revealed other affected relatives, unaffected carrier parents, and gonadal mosaicism. Individuals with germline mutations of SMARCB1 present at a younger age (5 vs. 18 months), may have multiple primary tumors, or affected siblings as expected with a germline tumor suppressor gene mutation [36, 38, 39].


Meningeal Tumors


Meningioma, a tumor that arises from membranes surrounding the brain, accounts for ∼20–30% of primary intracranial tumors. The World Health Organization classification of brain tumors recognizes three grades of meningioma: WHO grade I, the most common (70–80%); grade II, atypical (5–25%); and grade III (1–3%), the anaplastic type [1]. Grade I tumors are slow growing and benign with a low risk of recurrence, while grade II and III tumors have a greater likelihood of recurrence and/or aggressive behavior. Meningiomas occur most commonly in middle-age to older patients with a peak in the sixth and seventh decades. Female-to-male ratio is ∼2:1, with males overrepresented in grade II and III tumors [40, 41]. Pediatric (<20 years old) meningiomas are rare, comprising ∼2% of all meningiomas [42].

Meningioma was the first solid tumor to be associated with a nonrandom cytogenetic abnormality (monosomy 22). Loss of or interstitial deletion of chromosome 22q is the most common and often the sole anomaly in benign meningioma. Atypical and anaplastic meningiomas often show additional chromosome losses, i.e., 1p, 14q, 18, 10, and 6q. Spinal meningiomas are usually low grade with monosomy 22 as the sole anomaly [43, 44]. Pediatric meningiomas show similar chromosome anomalies as those in adults. A complex karyotype is more common in pediatric meningiomas, but there is insufficient data to correlate karyotype with biologic behavior in pediatric tumors [26, 42]. Mutations and/or deletions encompassing the NF2 gene at 22q12.2 are present in NF2-associated meningiomas and in ∼60% of sporadic meningiomas [1].



Genitourinary Tumors (Table 16.2)





Table 16.2
Chromosome abnormalities with diagnostic or clinical significance in genitourinary tumors















































































































































Tumor

Chromosomal aberration(s)

Gene(s) involved

Clinical significance

References

Renal

Clear cell RCC

Loss of 3 or 3p

VHL, PBRMI, PTH1R

Non-papillary RCC

[4548]

Loss of 3p with gain of 5q
 
Favorable prognosis

[4548]

Loss of 9p, 14q
 
Unfavorable, shorter survival

[4548]

Papillary RCC

Gain of 7, 17, loss of Y

MET

Adult papillary RCC

[45, 49]

t(Xp11.2) RCC

t(X;1)(p11.23;q23.1)

TFE3-PRCC

Xp11.2 RCCs infrequent in adult RCC, common in pediatric RCC

[5054]

t(X;17)(p11.23;q25.3)

TFE3-ASPSCR1

Balanced t(X;17) in RCC; Unbalanced t(X;17) in ASPS

[5054, 184]

t(X;17)(p11.23;q23.1)

TFE3-CLTC
 
[5054]

t(X;1)(p11.23;p34.3)

TFE3-SFPQ
 
[5054]

t(6;11) RCC

t(6;11)(p21.1;q13.1)

TFEB-ALPHA

Subset of RCC, children, young adults

[54, 55]

Chromophobe

Loss of 1, 2, 6, 10, 13, 17, 21
 
Distinguish from oncocytoma

[56, 57]

Oncocytoma

Loss of 1 or 1p, Y
 
Distinguish from chromophobe

[57]

Wilms tumor

Loss of 16q, 1p, 4q, 14q, 17p, 22; gain of 1q; der(1;16)(q10;p10)

TP53

Unfavorable histology; augmented chemotherapy if loss of 1p, 16q

[5864]

Clear cell sarcoma (CCSK)

t(10;17)(q22.3;p13.3), del(14)(q24.1q31.1)

FAM22-YWHAE

t(10;17) in 12% of CCSK; t(10;17) also seen in endometrial stromal sarcoma

[6568]

CMN

t(12;15)(p13.2;q25.3), gain of 11, 20, 17, 8

ETV6-NTRK3

Diagnostic; t(12;15) also seen in CFS/IFS and secretory breast cancer

[69, 70]

Rhabdoid tumor (RTK)

Loss of 22, del(22q)

SMARCB1-INBI1

Diagnostic

[7173]

Prostate

Gain of 7q, 8q, loss of 8p, 10q, 13q, 17p, 7p, 16q, 6q

MYC, PTEN, TP53, TMPRSS2-ERG

Poor OS with ERG amplification, TMPRSS2-ERG fusion+; poorest OS with PTEN-, TP53

[7580]

Bladder

Gain of 3, 5p, 7, 17, 20, loss of 9, 9p

CDKN2A

Homozygous deletion CDKN2A higher grade and stage; recurrence, progression

[8187]

Reproductive

Endometrial stromal tumor (EST)

t(7;17)(p15.2;q11.2), t(6;7)(p21.3;p15.2), t(6;10)(p21.3;p11.22), t(10;17)(q22.3;p13.3)

JAZF1-SUZ12, PHF1-JAZF1, PHF1-EPC1, FAM22-YWHAE

Distinguish from non-EST uterine tumors

[8891]

Germ cell

Postpubertal GCTs

12p overrepresentation, i(12p) or 12p amplification

12p

i(12p), amplification of 12p distinguishes GCTs

[95, 100104]

Ovarian dysgerminoma; testicular, seminoma, nonseminoma; extragonadal

12p overrepresentation, i(12p) or 12p amplification, gain of X, 7, 8, 12, 21, loss of 1p, 11, 13, 18

Chromosome enumeration

Mediastinal GCT associated with Klinefelter syndrome

[2, 7, 95, 98, 100, 102]

Prepubertal GCTs

Loss of 1p36, 4q, 6q; gain of 1q, 3p, 16p, 20q; rare gain of 12p

Chromosome enumeration

12p gain rare in prepubertal GCT

[96, 101, 102]


ASPS alveolar soft part sarcoma, CFS/IFS congenital fibrosarcoma/infantile fibrosarcoma, CMN congenital mesoblastic nephroma. CCSK clear cell sarcoma of kidney, GCT germ cell tumor, OS overall survival, RCC renal cell carcinoma, RTK rhabdoid tumor of kidney


Renal Cell Carcinoma


Renal cell carcinoma (RCC) is the most common malignant tumor arising from the kidney. Prognosis is related to histologic subtype and tumor stage at diagnosis. Histologic subtypes include clear cell RCC (70%), papillary RCC (10–15%), chromophobe RCC (4–6%), Xp11.2 translocation RCC, and others. Different subtypes are characterized by different genetic abnormalities. Hereditary syndromes with RCC as a feature include von Hippel-Lindau, Birt-Hogg-Dube, tuberous sclerosis, hereditary papillary RCC, familial clear cell RCC, hereditary leiomyomatosis and RCC, and familial oncocytoma. Hereditary RCCs account for 4% of RCCs [7].


Clear Cell RCC


Clear cell RCC (ccRCC) histology shows cells with clear or granular cytoplasm without a papillary growth pattern. The majority of ccRCCs have deletion or rearrangement of the short arm of chromosome 3 that results in loss of part or all of 3p, often including the von Hippel-Lindau (VHL) gene at 3p25.3. Mutations of VHL are present in ∼90% of sporadic tumors. No association between VHL status, tumor grade, and stage has been found. Other genes at 3p21.1 (PBRM1) and 3p21.31 (PTH1R) are reported to be mutated or lost in 40 and 76% of ccRCCs, respectively [4547]. In addition to 3p loss, ccRCCs may show gain of chromosome 5 or gain of 5q, gain of chromosome 7, and loss of 9p and/or 14q [48].

Recent work by Dondeti et al. further elucidated two subtypes of VHL deficient ccRCC—H1H2 and H2—based on HIF1A expression, with each group having its own specific pattern of copy number alterations. The H2 subtype showed gain of 5q and loss of 14q more frequently than H1H2, while the H1H2 subtype more frequently showed gain of 16p and 19p and loss of 6q. Gain of 5q, present in ∼30% of ccRCCs, is reported as a favorable prognostic factor, while losses of 9p and 14q are associated with a poor outcome [45]. Two genes identified on 5q—STC2 at 5q35.2 and VCAN at 5q14.3—are thought to be important in the tumorigenesis of ccRCCs [47].


Papillary RCC


Papillary RCC (pRCC) is characterized by a papillary growth pattern and occurs in familial and sporadic forms. Cytogenetically, pRCC shows gains of chromosomes 7 and 17 and loss of the Y chromosome (Fig. 16.6). Gains of chromosomes 12, 16, and 20 are also frequent. The MET proto-oncogene located at 7q31.2 is mutated in a subset of sporadic pRCC and is responsible for hereditary pRCC [45, 49].

A145460_3_En_16_Fig6_HTML.jpg


Fig. 16.6
Papillary renal cell carcinoma from the kidney of a 67-year-old male: 48,X,−Y,+7,+12,+17


TFE3 and TFEB Translocation RCC


RCC with Xp11.2 translocations/TFE3 gene fusions is seen in children and adults, but is more predominant in the pediatric age group. These tumors have a papillary architecture and resemble pRCC. Xp11.2 RCCs have been misclassified as chromophobe and ccRCCs.

Several partner genes fuse with TFE3 at Xp11.2. The two most common translocations are t(X;1)(p11.23;q23.1)/TFE3-PRCC and t(X;17)(p11.23;q25.3)/TFE3-ASPSCR1. Variants include t(X;1)(p11.23;p34.3)/TFE3-SFPQ and inv(X)(p11.23q13.1)/TFE3-NONO and others. The Xp11.2 translocation tumors are reported to be aggressive in both pediatric and adult patients, which may in part be due to higher stage disease at diagnosis [5054].

TFE3 is a member of the MiT transcription factor family. Another member, TFEB, is involved in a subset of RCCs. These RCCs show a t(6;11)(p21.1;q13.1). TFEB at 6p21.1 fuses with alpha at 11q13.1. Tumors with the t(6;11) are a distinctive subset of RCCs in children and young adults. The tumor has nests of epithelioid cells with clear cytoplasm along with a second population of smaller cells usually clustered around hyaline nodules [54, 55].


Chromophobe RCC and Oncocytoma


Chromophobe RCCs are composed of cells with prominent cell membranes and eosinophilic cytoplasm. They may be difficult to distinguish from the benign renal oncocytoma or the granular variant of ccRCC. Chromophobe RCC is characteristically hypodiploid with loss of multiple chromosomes including chromosomes 1, 2, 6, 10, 13, 17, and 21. Renal oncocytomas, composed of large eosinophilic cells, show a normal karyotype in 60% of tumors and partial or complete loss of chromosome 1 in 40%. Loss of the Y chromosome and chromosome 14 may be seen together with chromosome 1 loss [56, 57].


Wilms Tumor


Wilms tumor (WT) or nephroblastoma is the most common primary malignant renal tumor of childhood and the fourth most common pediatric malignancy overall. The classic WT is triphasic with blastemal, epithelial, and stromal components [2]. Most tumors are sporadic and unilateral.

Approximately 5–10% of patients with Wilms tumor have a germline predisposing gene mutation. WT is associated with congenital syndromes including Wilms tumor, aniridia, genitourinary anomalies, and mental retardation (WAGR) syndrome, Denys-Drash syndrome, Perlman’s syndrome, and Beckwith-Wiedemann syndrome. Patients with a germline mutation come to medical attention ∼10 months earlier than patients with sporadic tumors and may have bilateral tumors. With the treatment protocols of two large cooperative groups that prospectively study children with Wilms tumor (the National Wilms Tumor Study Group and the International Society of Paediatric Oncology), survival of patients with WT is now >85%. Prognosis correlates with histopathologic features; survival rates are lower with unfavorable histology (UFH) versus favorable histology (FH). Cytogenetic analysis and aCGH and LOH studies of Wilms tumor tissue have found that loss of 16q consistently correlates with UFH and higher mortality [58, 59]. Loss of 16q often results from an unbalanced der(1;16)(q10;p10). Deletions of 1p, 4q, 14q, 17p, and 22q and gain of 1q are associated with adverse outcome [6063]. Mutations of TP53 at 17p13.1 are associated with anaplasia, a feature of UFH tumors [63, 64].


Clear Cell Sarcoma of the Kidney


Clear cell sarcoma of the kidney (CCSK), the second most common pediatric renal tumor, is an unfavorable histology tumor with a propensity for recurrence and metastasis to bone, brain, and soft tissue. Little is known about the genetics of CCSK. Several individual cases have been reported with t(10;17)(q22.3;p13.3) and/or del(14)(q24.1q31.1) [65, 66]. O’Meara et al. recently identified the genes involved in the t(10;17) as FAM22 at 10q22.3 and YWHAE at 17p13. The translocation produces an in-frame fusion gene that is comprised of exons 1–5 of YWHAE and exons 2–7 of FAM22. Of 50 CCSKs studied by RT-PCR, only 12% were FAM22-YWHAE fusion positive [67]. This same translocation and gene fusion was also recently characterized and reported in high-grade endometrial stromal sarcomas [68].


Congenital Mesoblastic Nephroma


Congenital mesoblastic nephroma (CMN), the third most common pediatric renal tumor, is an uncommon spindle cell tumor diagnosed in infancy (60%) to <2 years of age. Infantile or congenital fibrosarcoma (CFS) is a soft tissue spindle cell tumor usually located in an extremity in children under 2 years old. Both CFS and CMN share a common translocation, t(12;15)(p13.2;q25.3), and gains, in decreasing frequency order, of chromosomes 11, 20, 17, and 8 (Fig. 16.7). t(12;15) fuses ETV6 with NTRK3. These chromosomal abnormalities distinguish CFS and CMN from other childhood spindle cell tumors, such as benign infantile fibromatosis or malignant adult-type fibrosarcoma [69, 70].

A145460_3_En_16_Fig7_HTML.jpg


Fig. 16.7
Cellular congenital mesoblastic nephroma, grade III, from the kidney of a 6-week-old boy: 48,XY,+11,+11,t(12;15)(p13.2;q25.3). This same t(12;15), which results in ETV6NTRK3 fusion, can also be seen in congenital/infantile fibrosarcoma and in secretory breast carcinoma (see text for details)


Rhabdoid Tumor of the Kidney


Rhabdoid tumor of the kidney (RTK), a neoplasm different from Wilms tumor, was given the name “rhabdoid” because microscopically it resembled a rhabdomyosarcoma (see later). RTK, a highly malignant neoplasm that occurs perinatally, during the first year of life, and occasionally in older individuals, is characterized by early metastases and a high mortality rate. Malignant rhabdoid tumors (MRT) occur in soft tissues, skin, CNS, and other extrarenal sites. Concomitant brain tumors are present in about one third of fetuses and neonates with RTK [71]. Loss of chromosome 22, 22q deletions, SMARCB1/INI1 mutations, and lack of INI1 immunostaining in histopathologic sections facilitate the diagnosis of MRTs [72, 73].


Prostate Cancer


Prostate adenocarcinoma is the most common cancer in males, representing 29% of cancers and causing 9% of cancer deaths in men [74]. Prognosis for patients with prostate cancer correlates with stage and grade of disease at diagnosis. Frequently reported chromosome alterations include loss of 8p, 10q (PTEN), 13q (RB1), 17p (TP53), 7p, 16q, 6q, and gain of 8q24.1 (MYC) and 7q31. Hemizygous or homozygous deletion of PTEN at 10q23.3 correlates with disease stage, disease progression, and survival [7577]. PTEN loss is among the most frequent recurring abnormalities in prostate cancer and is seen in preinvasive prostatic intraepithelial neoplasia as well as in invasive prostate cancer.

In 2005, TMPRSS2-ETS gene fusions were discovered in prostate cancer. TMPRSS2, transmembrane protease serine 2 gene, is androgen-regulated. The fusion of TMPRSS2 at 21q22.3 with an ETS gene results in overexpression of the 5′ truncated ETS oncogene. The most common fusion (TMPRSS2-ERG, present in >50% of prostate specific antigen-screened localized cancers) results from an interstitial ∼2.8 Mb deletion within 21q22; ERG is located at 21q22.2 [78, 79]. TMPRSS2-ERG fusion is associated with PTEN deletion and with earlier disease recurrence of localized prostate cancer after surgical resection. A study by Markert found that the poorest overall survival (57 months mean OS) correlated with TP53 and PTEN inactivation. TMPRSS2-ERG fusion positive tumors had a mean OS of 93 months compared with a mean OS of >103 months in the most favorable group. Data from this study and others suggests that a subset of TMPRSS2-ERG fusion-positive tumors with amplification of ERG have a poorer outcome [80].


Bladder Cancer


Bladder cancer can generally be divided into superficial, invasive, and in situ categories, which correspond to the TNM (“tumor, nodes, metastasized”) staging scheme. Tumor stage is the most important independent predictor of patient prognosis. Most tumors are superficial at diagnosis. However, during the course of the disease, multiple recurrences are common, with a minority of superficial tumors progressing to muscle invasion or metastatic disease. Identification of genetic changes in exfoliated cells from the bladder has shown utility in detecting disease recurrence. Primary noninvasive (Ta) or superficially invasive (T1) transitional cell carcinoma of the bladder can be monitored using FISH with centromeric and locus-specific DNA probes. UroVysion®, a US Food and Drug Administration (FDA)-approved FISH probe set, detects aneusomy of chromosomes 3, 7, and 17, and 9p21 (CDKN2A) loss in patients with hematuria or a history of bladder cancer [81, 82]. Recent studies support FISH analysis for use in monitoring and predicting recurrence risk in patients with non-muscle-invasive bladder cancer and predicting residual tumor load after transurethral resection [8386]. FISH analysis in combination with cytology and the telomeric repeat amplification protocol (TRAP) assay to detect telomerase activity increases the sensitivity of detection in low-grade and early-stage cancers [87]. See also Chap. 17, Fig. 17.16.


Tumors of the Reproductive System (Table 16.2)



Endometrial Stromal Tumor


Endometrial stromal tumors (EST) are rare uterine mesenchymal tumors that occur in women of reproductive and postmenopausal age. In 2003, the World Health Organization (WHO) divided ESTs into three categories: endometrial stromal nodules (ESN), endometrial stromal sarcomas (ESS), and undifferentiated endometrial sarcomas (UES). ESNs are benign, circumscribed tumors. ESSs are low-grade, malignant tumors that invade the myometrium. Both can have variant histologic features, but most have a classic morphology. UESs are highly aggressive, malignant tumors with cytologic atypia and high mitotic activity. ESSs may be primary uterine, metastatic uterine, or primary extrauterine. The tumors are characterized by reciprocal translocations and gene fusions with t(7;17)(p15.2;q11.2)/JAZF1-SUZ12 in ESN, ESS, and rarely UES, and t(6;7)(p12.3;p15.2)/JAZF1-PHF1 or t(6;10)(p12.3;p11.22)/EPC1-PHF1 and variants in ESS [8891]. Lee et al. recently identified a t(10;17)(q22.3;p13.3)/FAM22-YWHAE in high-grade ESS [68]. This same t(10;17) is found in clear cell sarcoma of the kidney (see earlier) [67].


Germ Cell Tumors (Table 16.2)


Germ cell tumors (GCTs) are a heterogeneous group of rare benign and malignant tumors. GCTs may arise in the gonads and at extragonadal sites found primarily in the body midline (intracranial, mediastinal, retroperitoneal, sacrococcygeal, and others). Primordial germ cells are thought to give rise to these tumors with aberrant germ cell migration responsible for the extragonadal tumors. While the tumors may arise from the same cell type, the clinical course and outcome of the various GCTs differ depending on tumor site and histology [9294]. Tumors are found prenatally into old age with diverse groups: neonates, infants and children ≤5 years of age, postpubertal to the fifth decade, and older age.


Gonadal Germ Cell Tumors


Testicular GCTs (TGCT), divided into seminomas and nonseminomas, are the most common tumor of men in the second to fourth decades and are responsible for ∼10% of cancer deaths of men in this age bracket. The US incidence is ∼6/100,000 with a 5:1 white: black ratio. Cryptorchidism is associated with ∼10% of TGCTs. Seminomatous GCTs, ∼40–50% of GCTs, are composed of cells that resemble primordial germ cells. Nonseminomatous GCTs (NSGCT) may be composed of undifferentiated cells that resemble embryonic stem cells. Malignant cells can differentiate to generate yolk sac (endodermal sinus) tumor, embryonal carcinoma, choriocarcinoma, teratomas, and mixed malignant GCTs. Approximately 60% of TGCTs are composed of more than one of these cell types [95]. TGCT of infants are rare neoplasms occurring in boys 0–4 years of age, the majority of which are pure yolk sac tumors [96].

GCTs account for 15–20% of all ovarian tumors. Most are benign cystic teratomas. The remaining, primarily found in children and young adults, may be malignant with histologic types similar to those in the testis. The counterpart to the testicular seminoma is the ovarian dysgerminoma, which is always malignant. Dysgerminomas account for ∼2% of all ovarian tumors, but ∼50% of GCTs. They may occur in patients with gonadal dysgenesis.


Extragonadal Germ Cell Tumors


Rare primary brain GCTs occur in the midline, pineal (male predominance), or suprasellar regions. They account for 0.2–1% of brain tumors in those of European descent, but up to 10% in those of Japanese origin.

Primary mediastinal germ cell tumors account for 10–15% of all mediastinal tumors [97]. Pediatric mediastinal GCTs represent ∼5% of all GCTs [98]. These tumors have the same histologies as gonadal GCTs, but have a worse prognosis. The mediastinum is the most common site of extragonadal tumors in young males. Mediastinal NSGCTs are associated with Klinefelter syndrome in ∼20% of cases [7]. Patients with Klinefelter syndrome have a 50-fold higher risk for a mediastinal GCT, but do not develop testicular GCT [2].

Sacrococcygeal GCTs, the most common extragonadal GCT in children, present prenatally to ∼4 years of age. Most (∼90%) external lesions are benign, while intrapelvic or intra-abdominal tumors are more likely to be malignant (60–90%) [99].


Chromosome Abnormalities in Germ Cell Tumors


Cytogenetically, additional copies of all or part of 12p are the characteristic chromosome abnormality associated with GCTs in adults and postpubertal children [95, 100]. Additional copies of 12p are present as i(12p) in 80–90% of cases, while the remaining tumors show 12p amplification. Additional cytogenetic anomalies in this group include gains of an X chromosome and chromosomes 7, 8, 12, and 21 and loss of 1p, 11, 13, and 18 (Fig. 16.8).

A145460_3_En_16_Fig8_HTML.jpg


Fig. 16.8
Malignant mixed germ cell tumor from the mediastinum of an 8-year-old boy: 96<4n>,XXYY,+X,+X,+1,idic(1)(p22),−4,−5,−7,+i(12)(p10)x4,+21,+21,−22. In addition to other abnormalities, this karyotype results in gain of 12p in the form of isochromosomes for the short arm (lower left) (see text for details)

GCTs in infants and prepubertal children characteristically show loss of 1p36, 4q, and 6q, and gain of 1q, 3p, 16p, and 20q. Gain of 12p is rarely reported in prepubertal children and infants [101, 102]. Array CGH has shown proximal 12p11.2-p12.1 gain associated with adult TGCTs and distal 12p12-pter gain in yolk sac tumors of very young children [102104]. GCTs are rare in children between 5 and 9 years of age; thus there is a paucity of tumor genetic information in this age range.


Gastrointestinal Tumors (Table 16.3)





Table 16.3
Chromosome abnormalities with diagnostic or clinical significance in gastrointestinal tumors




















































































































Tumor

Chromosomal aberration(s)

Gene(s) involved

Clinical significance

References

GIST

Loss of 14q, 22q, 1p, 15q, 13q; gain of 1q, 12q

KIT, PDGFRA

KIT, PDGFRA mutation diagnostic, response to TKIs

[105111]

Liver

Hepatoblastoma

Gain of 1q, 2, 8, 20, der(4)t(1;4), loss of 4q
 
Distinguish from HCC, HMH

[112, 113]

Hepatic mesenchymal hamartoma (HMH)

t(11;19)(q13;q13.4), t(19q13.4)

Unknown genes

Distinguish from hemangioma or malignant tumor

[120]

Salivary gland

Pleomorphic adenoma

t(3;8)(p22.1;q12.1), t(12q14.3), +8

CTNNB1-PLAG1, HMGA2

Diagnostic; benign tumor

[127129]

Ca-ex-PA

HMGA2, MDM2 amplification

HMGA2, MDM2

Amplification contributes to malignant transformation of PA

[128]

Mucoepidermoid cancer

t(11;19)(q21;p13.11) in 40–80%; gain of 7, 8, X, loss of 6q

MAML2-CRTC1

Malignant; t(11;19) assoc with better outcome

[130, 131]

Warthin’s tumor

t(11;19)(q21;p13.11) in low percentage

MAML2-CRTC1

Benign; t(11;19) w/metaplasia

[132]

Dermal

DFSP and variants (GCF, Bednar, other)

t(17;22)(q22;q13.1), der(22)t(17;22) or r(22)t(17;22)

COL1A1-PDGFB

Diagnostic for DFSP; response to TKIs

[135140]

Hidradenoma

t(11;19)(q21;p13.11), gain of 7, 8, X, loss of 6q

MAML2-CRTC1

Clear cell variant

[133, 134]

Cutaneous melanoma

Gain of 6p, 1q, 7, 8q, 17q, 11q, 20q; loss of 9, 9p, 10q, 6q

CDKN2A, BRAF, PTEN

CDKN2A

[143, 144]

Uveal melanoma

Loss of 3, gain of 8q

GNA11, GNAQ

Monosomy 3 correlates with metastatic disease

[145, 146]

Breast

Invasive intraductal

dmin, hsr (ERBB2 amplification)

ERBB2

Improved outcome with targeted therapy

[149151]

Secretory breast

t(12;15)(p13.2;q25.3)

ETV6-NTRK3

Favorable; distinguish from other breast lesions

[152]

Lung

NSCLC

EGFR high copy number or amplification, loss of 3p, gain of 7

EGFR

Response to TKIs

[155]
 
inv(2)(p21p23.2)

EML4/ALK

Response to TKIs

[156, 157]


Ca-ex-PA Carcinoma ex Pleomorphic Adenoma, DFSP dermatofibrosarcoma protuberans, dmin double minutes, GCF giant cell fibroblastoma, GIST gastrointestinal tumor, HCC hepatocellular carcinoma, hsr homogeneously stained regions, NSCLC non-small cell lung carcinoma, TKI tyrosine kinase inhibitor


Gastrointestinal Stromal Tumors


Gastrointestinal stromal tumors (GIST), the most common mesenchymal tumor of the gastrointestinal (GI) tract, arise from the connective tissue of the GI wall. Approximately 90% of GISTs have activating mutations of the KIT or PDGFRA receptor tyrosine kinase genes [105]. In addition to KIT or PDGFRA mutations, other genetic events involved in tumorigenesis include primarily chromosomal losses (14q, 22q, 1p, 15q, 13q), nuclear/mitochondrial microsatellite instability, LOH at 9p21 (CDKN2A), methylation of CDKN2B (p15), homozygous loss of TLX2 (HOX11L1), and rare gene amplification (MYC, MDM2, EGFR, CCND1, KIT) [106109]. The KIT and PDGFRA mutations are diagnostic, and tyrosine kinase inhibitors (TKIs) are used as targeted therapy. The molecular aberrations of KIT-PDGFRA are correlated with cell histomorphology, metastasis, prognosis, and efficacy of targeted therapy. Genotyping these tumors helps to guide therapy, as the effects of TKIs vary with the presence or absence and site of KIT-PDGFRA mutation. Metastatic GISTs often have secondary KIT kinase mutations, and some have KIT-PDGFRA genomic amplifications, which are responsible for therapeutic resistance [105]. Disease-free survival correlates with mutation (KIT  =  poor), site (stomach  =  best), cytogenetic complexity (≥3 abnormalities  =  poor), and losses of 1p and/or 22q and gains of 1q and 12q (shorter survival) [110, 111]. GISTs in patients with neuro-fibromatosis type I (NF1) lack KIT and PDGFRA mutations. Rare families have been reported with germline KIT or PDGFRA mutations [105, 110].


Liver Tumors



Hepatoblastoma


Hepatoblastoma (HB) is the most common primary malignant tumor of the liver in children. This rare tumor accounts for ∼1% of all pediatric malignancies, with ∼100–150 new tumors per year in the United States.

Cytogenetic analysis of hepatoblastomas has found that the most common anomalies are numerical, with gain of whole chromosomes, specifically of chromosomes 20, 2, and 8, in decreasing order of frequency (Fig. 16.9). The most common structural abnormalities result in gain of chromosome 1 long-arm material. An unbalanced der(4)t(1;4) that results in gain of 1q and loss of 4q is the most common recurring structural abnormality [112]. Rare genomic and expression profiling studies have confirmed these abnormalities and further refined the regions of gain and loss. Single nucleotide polymorphism (SNP) array analysis revealed paternal 11p uniparental disomy (UPD, see Chap. 20) that included the IGF2-H19 region at 11p15.5 [113]. Molecular studies have discovered mutations in key genes that are important in the genetic pathways of the developing liver [114]. These studies may help elucidate the pathogenetic mechanisms responsible for the development of hepatoblastoma. SMARCB1 (INI1) testing helps differentiate hepatoblastoma from a more aggressive variant that mimics rhabdoid tumor.

A145460_3_En_16_Fig9_HTML.jpg


Fig. 16.9
Hepatoblastoma, mixed embryonal and fetal, from the liver of a 9-year-old extremely premature boy: 50,XY,del(1)(q32q42),add(2)(q23),+add(2)(q31),add(3)(p21),add(6)(q23),+8,+12,add(14)(q13),+20

Most cases of HB are sporadic. However, HB is associated with several cancer predisposition syndromes including Beckwith-Wiedemann syndrome (BWS), familial adenomatous polyposis, and Li-Fraumeni syndrome. HB can also be seen in glycogen storage disease type I. Premature infants, particularly those with low or very low birth weight, are at increased risk of developing hepatoblastoma [115].

Familial adenomatous polyposis (FAP), a syndrome of early-onset colonic polyps and adenocarcinoma, results from germline mutations in the APC tumor suppressor gene at 5q22.2. Children with a family history of FAP have a significantly increased risk for hepatoblastoma [116]. One study estimated that 1 in 20 hepatoblastomas is probably associated with FAP. APC mutations, common in patients with hepatoblastoma and FAP, are rare in patients with sporadic hepatoblastomas [117, 118]. Children who survive HB should be considered for evaluation of FAP, and those patients found to carry an APC mutation need close surveillance because of their increased risk for colonic polyps and adenocarcinoma.

Patients with hemihypertrophy or Beckwith-Wiedemann syndrome should be screened using α-fetoprotein (AFP) as a marker to detect hepatoblastoma. AFP monitoring should be performed every 3 months until the child is at least 4 years. Loss of heterozygosity of 11p markers occurs commonly in hepatoblastoma associated with BWS and hemihypertrophy.

Children with HB often have very elevated AFP levels and may have anemia and thrombocythemia. Complete surgical removal effects a cure. However, ∼70% of tumors are metastatic or unresectable at diagnosis. Even with aggressive chemotherapy, 25–30% remain resistant. AFP levels return to normal with tumor removal and rise if the tumor returns, thus providing a monitor for disease [115, 119].


Hepatic Mesenchymal Hamartoma


Mesenchymal hamartoma of the liver (HMH) is a rare benign lesion that occurs mainly in infants. Histologically, the lesion has cystic and solid areas with islands of hepatocytes and poorly defined or dilated biliary ducts in a myxoid stroma. Complete surgical removal is curative. In the few reported cases, a common denominator has been involvement of chromosome band 19q13.4 with t(11;19)(q13;q13.4) so far the most common reciprocal translocation [120].

HMH has been associated with placental mesenchymal dysplasia (PMD), an uncommon disorder of the placenta. Placental changes include cystic villi with dilated/thick-walled vessels, which can mimic a partial hydatidiform mole. In contrast to a partial mole, PMD can coexist with a normal viable fetus [121]. Both HMH and PMD have been considered developmental aberrations rather than true neoplasms [120].

Further analysis suggests PMD may be a disease of dysregulated imprinting with mosaic placental and fetal paternal UPD [122124]. HMH and PMD have been associated with Beckwith-Wiedemann syndrome (BWS), an imprinting disorder [125]. One cause of BWS is paternal UPD at 11p15.5. A paternally imprinted gene, PEG3, is within the locus at 19q13.43 that is commonly disrupted in HMH [123].

While HMH is considered to be benign, there is a low risk of malignant transformation. Several malignant tumors of undifferentiated embryonal sarcoma (UES) of the liver have been reported to arise from HMH. Cases of UES have been reported to have involvement of the same 19q13.4 locus as that of HMH [120, 126].


Salivary Gland Tumors



Pleomorphic Adenoma


Pleomorphic adenoma (PA), a benign mixed salivary gland tumor, has been associated with abnormal karyotypes in the majority of cases, with nonrandom involvement of 8q12.1 (locus of the pleomorphic adenoma (PLAG1) gene), 3p22.1 (CTNNB1), 12q14.3 (HMGA2), and gain of chromosome 8 [127, 128]. t(3;8)(p22.1;q12.1) is the most common translocation (Fig. 16.10). Reported partner genes for PLAG1, CTNNB1, and HMGA2 vary.

A145460_3_En_16_Fig10_HTML.jpg


Fig. 16.10
Pleomorphic adenoma, submandibular, from a 10-year-old girl: 46,XX,t(3;8)(p22.1;q12.1). The translocation results in CTNNB1-PLAG1 fusion

Few PAs (∼6%) undergo malignant transformation to carcinoma ex pleomorphic adenoma (Ca-ex-PA). Recent work has shown that HMGA2 translocations are often associated with gene amplification. There is an increased risk of malignant transformation with HGMA2 amplification. HGMA2 is usually co-amplified with others genes, most often MDM2.

Other genetic abnormalities thought to contribute to malignant transformation of PA to Ca-ex-PA include deletions of 5q23.2-q31.2 and TP53, gains of 8q12.1 (PLAG1) and 8q22.1-q24.1 (MYC), and ERBB2 amplification [129].


Mucoepidermoid Carcinoma


Mucoepidermoid carcinoma (MEC) accounts for ∼15% of salivary gland tumors and is the most common primary malignant tumor of the salivary gland. MEC arises predominantly, but not exclusively in the parotid gland. A t(11;19)(q21;p13.11) that results in CRTC1-MAML2 fusion is found in 40–80% of tumors (Fig. 16.11). Evidence from several studies found that fusion positive tumors are less likely to recur or metastasize and are associated with an overall better survival [130, 131]. t(11;19) negative cases show gain of chromosomes 7, 8, and X and 6q deletion [131].

A145460_3_En_16_Fig11_HTML.jpg


Fig. 16.11
Mucoepidermoid carcinoma from the parotid gland of a 9-year-old boy: 46,XY,t(11;19)(q21;p13.11). This same t(11;19), which results in MAML2-CRTC1 fusion, can be seen in hidradenoma, a benign sweat gland tumor


Warthin’s Tumor


Warthin’s tumor, the second most common salivary gland tumor, is a benign neoplasm that arises almost exclusively in the parotid gland. Warthin’s tumor, also referred to as papillary cystadenoma lymphomatosum, is composed of polyclonal lymphoid cells and neoplastic epithelium. Recurrence and malignant transformation occur very rarely [2]. t(11;19)(q21;p13.11), which results in CRTC1-MAML2 fusion, is found in a low percentage of Warthin’s tumors [132]. Tumors that exhibit the translocation or are fusion-positive characteristically have metaplasia of the oncocytic epithelium. There is ongoing discussion regarding the association of the t(11;19) CRTC1-MAML2 fusion with both Warthin’s tumor and MEC; there is morphologic overlap between metaplastic Warthin’s tumor and MEC [130]. Further, clear cell hidradenoma, a benign sweat gland tumor, also demonstrates the t(11;19) and CRTC1-MAML2 fusion [133, 134].


Dermal Tumors (Table 16.3)



Dermatofibrosarcoma Protuberans


Dermatofibrosarcoma protuberans (DFSP) is an intermediate-grade soft tissue malignancy that usually arises in the dermis and subcutaneous tissue of adults and rarely in children [135]. DFSP is a slow-growing infiltrative dermal neoplasm with a propensity to recur locally after surgical resection, but is rarely metastatic (1–4%). Tumor-related deaths are very rare. There are several histologic variants of DFSP, such as giant cell fibroblastoma, Bednar tumor, and other fibrohistiocytic tumors, which should be considered in the differential diagnosis.

DFSP is characterized by a balanced or unbalanced form of a translocation t(17;22)(q21.3;q13.1) or by supernumerary ring chromosomes derived from this chromosome 22 [der(22)r(17;22)] that contain low-level amplified sequences from 17q21.31-qter and from 22q11.1-q13.1 [136]. The unbalanced form is usually a der(22)t(17;22) (Fig. 16.12). Both the ring and linear forms of the derivative chromosome 22 result in fusion of the α(alpha)-1 chain of type 1 collagen gene (COL1A1) at chromosome 17q21.3 with the second exon of the platelet-derived growth factor–β (PDGFB) gene at chromosome 22q13.1. Variability of the COL1A1 break point has no correlation with any clinical or histological parameter [135, 137139]. However, cytogenetically, ring chromosomes are common in adult DFSP, while the translocation derivatives are seen in all childhood cases [140]. The COL1A1-PDGFB chimeric gene protein causes unregulated expression of platelet-derived growth factor leading to abnormal activation of the platelet-derived growth factor receptor beta (PDGFRB) tyrosine kinase through an autocrine loop. Tyrosine kinase inhibitors are the current therapy for recurrent, metastatic, or inoperable tumors [141, 142]. Demonstration of the COL1A1-PDGFB fusion is necessary for the diagnosis of DFSP or DFSP variants [137].

A145460_3_En_16_Fig12_HTML.jpg


Fig. 16.12
Dermatofibrosarcoma protuberans from the breast of a 2-year-old girl: 50,XX,+4,+11,+18,+der(22)t(17;22)(q21.3;q13.1). The arrow indicates the chromosome 22 derived from the translocation, which results in COL1A1-PDGFB fusion (see text for details)


Variants of DFSP


Several histologic variants of DFSP are described including giant cell fibroblastoma (GCF), pigmented Bednar tumor (BT), DFSP with fibrosarcoma (FS)-like changes (DFSP-FS), and others.

Giant cell fibroblastoma (GCF), also called juvenile DFSP, more commonly affects infants and children. Bednar tumor, a pigmented form of DFSP with additional melanin-containing dendritic cells, occurs in early to middle adulthood. DFSP-FS is a more cellular form with higher mitotic rate. All variants are characterized by COL1A1-PDGFB fusion [137, 140].


Hidradenoma


Hidradenoma, a benign sweat gland tumor, often presents as a solitary, slow-growing, solid, or cystic intradermal nodule. Malignant transformation is uncommon. t(11;19)(q21;p13.11) is characteristic of a subset of these tumors (Fig. 16.11). The CRTC1-MAML2 fusion was demonstrated in 50%, specifically in tumors with clear cells, representing the clear cell variant of hidradenoma [133]. As noted earlier, salivary gland Warthin’s tumor and mucoepidermoid carcinoma also demonstrate the t(11;19)/CRTC1-MAML2 fusion. The common glandular denominator for these different tumors suggests they may originate from a common progenitor cell in salivary, bronchial, and sweat glands [130, 133].


Malignant Melanoma


Melanomas are malignant lesions, primarily cutaneous, but may occur on mucosal surfaces and in the eye. Cutaneous malignant melanoma, a pigmented skin lesion, may be lethal if not recognized and completely excised prior to metastasizing. Malignant melanomas spread superficially before progressing to invade the deeper dermal tissues. Malignant melanoma may be recognized by visible changes of a pigmented lesion (mole) characterized by changes in size and color and irregular borders, and may evolve from dysplastic nevi. Individuals with dysplastic nevus syndrome have a 50% risk for developing melanoma by 60 years of age. Frequent aberrations found in melanomas include gains of 6p, 1q, 7p, 7q (BRAF), 8q, 17q, 11q, and 20q and losses of 9p (CDKN2A), 9q, 10q (PTEN), 10p, and 6q [143, 144].

Uveal melanoma, the most common form of primary eye cancer, is characterized by loss of chromosome 3 in ∼50% of tumors; metastasis is correlated with such loss. Chromosome 3 loss is often accompanied by i(8q); tumors without loss of chromosome 3 have 6p abnormalities.

Two regions of chromosome 3, 3p25 and 3q24-26, appear to harbor tumor suppressor genes. More than 80% of uveal melanomas have been found to have a constitutively active somatic mutation of one of two genes, GNA11 at 19p13.3 and/or GNAQ at 9q21.2. These genes appear to contribute to the development of these tumors [145, 146].


Epithelial Cancer (Table 16.3)



Breast Cancer


Breast carcinoma is the most common cancer in women and the second leading cause of cancer deaths in women. The lifetime risk for breast cancer for women in the general population is 1 in 8. A positive family history of breast cancer increases this risk. A germline mutation of one of the known breast cancer predisposing genes greatly increases risk [2]. Currently known breast cancer genes explain only ∼30% of the heritability. Mutations of the breast cancer predisposing genes BRCA1 and BRCA2 account for ∼16–20% of the familial risk of breast cancer in the general population [147, 148].


Invasive or Metastatic Breast Cancer


Prognosis for patients with breast cancer correlates with stage, histologic type and grade, hormonal (estrogen and progesterone) receptor, and ERBB2 (HER2) status. Amplification and/or protein overexpression of ERBB2, found in ∼20% of new diagnosis breast cancer, is associated with more aggressive disease and decreased survival time. Accurate assessment of ERBB2 oncogene status is critical to care of the patient with invasive or metastatic breast cancer as it is used in selection of therapy. The risk of recurrence and mortality are reduced by ∼50% and ∼33%, respectively, in patients with early-stage ERBB2-positive tumors treated with trastuzumab (Herceptin®). Data indicate that patients with tumors that show ERBB2 overexpression (3+ by IHC) or gene amplification (by FISH) be considered a candidate for anti-ERBB2 therapy [7] (see also Chap. 17, Fig. 17.15). Because correlation between the IHC and FISH is <100%, guideline recommendations for ERBB2 testing were established by an expert panel of members from the American Society of Clinical Oncology (ASCO) and the College of American Pathologists (CAP) [149151].


Secretory Breast Carcinoma


Secretory breast carcinoma (SBC), a rare subtype of breast cancer, is characterized by abundant eosinophilic secretions in intracellular vacuoles and intercellular spaces. SBC occurs in both sexes and in children and adults, but is most often seen in young adult females. Most tumors are hormone receptor and ERBB2 negative. SBC is associated with a favorable prognosis, even in cases with local recurrence or ≤3 positive lymph nodes. SBC is characterized by t(12;15)(p13;q26)/ETV6-NTRK3 fusion, which results in a chimeric tyrosine kinase fusion product. This same t(12;15) ETV6-NTRK3 fusion is also seen in congenital (infantile) fibrosarcoma and congenital cellular mesoblastic nephroma [152] (see Fig. 16.7).


Lung Cancer


Lung cancer, the most common cancer worldwide, is largely due to tobacco products. Incidence and mortality rates of lung cancer have been declining since ∼1990 secondary to decreased smoking rates over the past 30 years. The most common types are non-small cell carcinomas (which include adenocarcinoma, squamous cell carcinoma, and large-cell carcinoma) and small-cell carcinomas [7].


Non-small Cell Lung Carcinoma


Adenocarcinoma is the most common type of lung cancer in women and nonsmokers. KRAS mutations occur primarily in adenocarcinoma, while TP53, RB1, and CDKN2A mutations occur in squamous cell and adenocarcinoma. EGFR mutations and amplification occur more frequently in patients with adenocarcinoma histology, no history of smoking, East Asian ethnicity, and female gender [2].

Up to 20% of non-small cell lung carcinomas (NSCLC) have EGFR mutations and/or amplification, and ∼80–85% of patients with EGFR mutations respond to therapy with tyrosine kinase inhibitors. FISH analysis detects EGFR amplification (defined as a gene:chromosome ratio ≥2, or ≥15 copies per cell in ≥10% cells) and polysomy (defined as ≥4 copies in ≥40% of cells) [153, 154]. Fukuoka et al. reported that EGFR mutation was the strongest predictive biomarker for benefit of gefitinib over carboplatin/paclitaxel on progression-free survival (PFS) and overall response rate (ORR). PFS was significantly longer with gefitinib for patients whose tumors had both high EGFR gene copy number and EGFR mutation [155].

A subset of NSCLCs exhibit ALK gene rearrangement, e.g., EML4-ALK, which results from an inv(2)(p21p23.2). The EML4-ALK fusion is found predominantly in younger (average 52 years) nonsmokers with adenocarcinoma histology. The fusion protein causes constitutive activation of the ALK tyrosine kinase. Most reports show no overlap with EGFR or RAS gene mutations. ALK positive patients have shown significantly better overall survival at 1 and 2 years when treated with crizotinib, a drug targeted against the constitutively active tyrosine kinase. The FDA has approved treatment with crizotinib with FISH testing as a companion diagnostic test for ALK detection [156, 157]. See also Chap. 17.


Bone and Soft Tissue Tumors (Table 16.4)





Table 16.4
Chromosome abnormalities with diagnostic or clinical significance in bone and soft tissue tumors


















Tumor

Chromosomal aberration(s)

Gene(s) involved

Clinical significance

References

CFS/IFS

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jun 17, 2017 | Posted by in BIOCHEMISTRY | Comments Off on The Cytogenetics of Solid Tumors

Full access? Get Clinical Tree

Get Clinical Tree app for offline access