Basic Concepts in Head & Neck Surgery and Oncology Krishnakumar Thankappan, Subramania Iyer
INDEX
×
Chapter Notes

Save Clear


1Thyroid
  • • Pathology of Thyroid Tumors
  • • Guidelines in the Management of Differentiated Thyroid Cancer
  • • Guidelines in the Management of Medullary and Anaplastic Thyroid Cancer
  • • Principles of Thyroid Surgery
  • • Role of Nuclear Medicine in Adjuvant Therapy and Follow-up of Differentiated Thyroid Cancer
  • • Medical Management of Differentiated Thyroid Cancer2

Pathology of Thyroid TumorsCHAPTER 1

Smitha NV
 
Introduction
Thyroid tumors account to 1% of all malignancies in developed countries and 0.2% of cancer deaths. They are the most common malignancies of the endocrine system and pose a significant challenge to pathologists, surgeons and oncologists. Most of the carcinomas affect young and middle aged adults and are indolent malignancies with a 10 year survival that exceeds 90%. There has been an increase in the incidence rate of these tumors worldwide which can be largely attributed to more sophisticated diagnostic methods and a change in diagnostic practices with an increasing number of smaller tumors being detected of late. Thyroid tumor pathology is an area replete with diagnostic challenges. Though there are typical morphological patterns described, overlaps with non-neoplastic entities pose diagnostic difficulties. Updates in this field include ancillary and research aiming at techniques that can further narrow down our diagnosis from the different ‘indeterminate/grey zone’ lesions detected on screening.
 
Classification
The conventional classification based on morphology and clinical features is largely supported by molecular data currently available. Genetic profiles of four main categories appear distinctly different from each other with a few areas of overlap.
The classification of thyroid tumors modified from WHO classification (2004) is as follows:
 
Tumors of Follicular Epithelium
  • Follicular adenoma (including Hurthle cell adenoma)
  • Hyalinizing trabecular adenoma
  • Follicular carcinoma (including Hurthle cell carcinoma)
    • Minimally invasive
    • Widely invasive
  • Papillary carcinomas
  • Poorly differentiated carcinoma
  • Anaplastic carcinoma
  • Squamous cell carcinoma
  • Mucoepidermoid carcinoma
  • Sclerosing Mucoepidermoid carcinoma with eosinophilia
  • Mucinous carcinoma.
 
Tumors with C Cell Differentiation
  • Medullary carcinoma.
 
Tumors with Mixed Differentiation
  • Collision tumor—follicular/papillary or follicular/medullary
  • 4Mixed differentiated carcinoma intermediate type.
 
Tumors showing Thymic or Related Branchial Pouch Differentiation
  • Ectopic thymoma
  • Spindle epithelial tumor with thymus-like element (SETTLE)
  • Carcinoma showing thymus-like element (CASTLE).
 
Tumors of Lymphoid Cells
  • Malignant lymphoma
  • Plasmacytoma.
 
Mesenchymal Tumors
  • Smooth muscle tumors
  • Peripheral nerve sheath tumors
  • Paragangliomas
  • Solitary fibrous tumors
  • Follicular dendritic cell tumors
  • LCH
  • Angiosarcoma.
 
Teratomas
 
Secondaries
 
Cytology of Thyroid Tumors
Fine needle aspiration (FNA) still remains the mainstay for screening and diagnosis of thyroid lesions.
Category
Risk of malignancy (%)
Management
I
ND (Nondiagnostic)
1–4
Repeat FNA with US
II
Benign
<1
Follow-up
III
AUS (Atypia of undetermined significance)
Approx. 5–10
Repeat FNA
IV
Suspicious for a Follicular Neoplasm (FN)
15–30
Lobectomy
Suspicious for a Hurthle cell Neoplasm (HCN)
15–45
V
Suspicious for malignancy
60–75
Lobectomy or Total thyroidectomy
VI
Malignant
97–99
Total thyroidectomy
There are several reporting systems which are being used to guide further management of the lesions. Most widely used one is that of Bethesda system. The system divides the lesions in to six diagnostic categories.
 
Bethesda System
Borderline lesions (category 3) make up to 10–15% of the cases. Overlapping cytological features of thyroiditis with papillary carcinomas and cellular colloid nodules with follicular neoplasms are particularly troublesome.
 
FNA Thyroid
  • Sensitivity—65–98%
  • Specificity—50–96%
  • False negativity—1–11%
  • False positivity—0–7%.
Studies have shown that 2–37% of unsatisfactory samples show malignancy in subsequent excisions, thus, stressing the need for excising lesions which yield repeatedly bloody aspirates. FNA can also be used to detect metastatic malignancies. Newer diagnostic methods in cytology material are being devised to check for mutations (BRAF) at a molecular level which will be of aid in borderline lesions.
 
Papillary Carcinoma (Figs 1.1 and 1.2)
This is the most common malignant tumor of the thyroid gland and comprises 80–85% of all 5malignancies. It is common in countries having iodine sufficient or iodine excess diets.
zoom view
Fig. 1.1: Papillary thyroid carcinoma showing cells in complex and branching papillae (H & E, 2X)
They tend to be biologically indolent and have an excellent prognosis. Papillary carcinoma can occur at any age, but most of them are diagnosed in third and fifth decades of life. Women are more frequently affected (2–4: 1). Multifocality is common. Most important etiological role is that of radiation. It was frequently diagnosed in patients who are treated with low dose radiation to head and neck for benign diseases. It was also recognized in survivors of atomic bomb explosion in Japan. Survivors of other cancers who were treated with radiation were also found to develop papillary carcinomas as second primaries. Dietary iodine concentration appears to influence incidence and in some cases, the morphology of papillary carcinomas.
 
Pathology
Papillary carcinoma shows varied gross features. Majority of the cases present as a solid irregular and firm grey white growth with granular cut surface. Scarring may be very prominent in some of the cases. Calcification is a common finding. It can also present as a small scar in the subcapsular location. Some lesions may be completely cystic with a solid mural nodule attached.
zoom view
Fig. 1.2: Papillary carcinoma with overlapping clear ‘Orphan Annie nuclei’ with nuclear grooves and inclusions (H & E, 40X)
PTC is characterized by unique nuclear features which are diagnostic of this entity. Typical cytological findings include cells in papillae with anatomical borders and monolayered sheets showing swirling. They have ovoid, overlapping nuclei with grooves and intranuclear inclusions. Studies have shown that presence of a combination of papillae, intranuclear inclusions and metaplastic squamoid cytoplasm is 98% predictive of papillary carcinoma in cytology material. Nuclear grooves are also seen in thyroiditis, HTA and adenomatous hyperplasias while intranuclear inclusions are encountered in medullary carcinomas, HTAs and paragangliomas. Histological sections show overlapping and clearing of nuclei typically described as Orphan Annie nuclei.
Depending on different patterns, cell types and clinical features, different variants have been described. Except for oncocytic and hobnail variants, all others should have more than 50% of the tumors showing their unique patterns along with nuclear features for their diagnosis. An oncocytic variant shows oncocytic morphology in ≥75%; hobnail variant shows hobnailed nuclei in ≥30%.
Among the variants, specific prognostic significance is connoted to the following types:
  • 6Tall cell variant
    • Cells should be 3 times taller than their width
    • Accounts for 10% of PTC cases
    • More seen in elderly
    • Usually large and show extrathyroid extension and recurrences more frequently
    • 10 year survival rate is 70%
    • Less sensitive to RAI therapy
    • High prevalence of BRAF mutation.
  • Diffuse sclerosing variant
    • More seen in children and young adults
    • Clinically aggressive
    • All cases are associated with lymph node metastasis at the time of presentation
    • Diffusely infiltrating tumor with sclerotic stroma showing squamous morules, psammoma bodies and associated thyroiditis
    • Lung metastasis is also more common at presentation (25%).
  • Columnar cell variant
    • Very rare
    • High columnar cells with pseudostratification, supra and subnuclear vaccuolations reminiscent of early secretory endometrium
    • Clinically aggressive.
  • Solid variant
    • Mimics solid type of poorly differentiated carcinoma, but do not have its guarded prognosis.
  • Cribriform morular variant
    • Seen typically in patients of FAP and Gardners syndrome (APC mutations)
    • Shows cribriform features, solid and spindle areas with squamoid morphology
    • Usually multifocal.
  • Clear cell variant
    • Recognition of this variant at metastatic sites can be problematic without immunostains.
  • Hobnail variant
    • Recently described entity
    • Clinically aggressive.
  • Oncocytic variant
    • Resistant to RAI therapy
    • Clinically aggressive.
  • Follicular variant
    • Unique variant with different genetic profile
    • Often diagnosed as follicular neoplasms in FNA due to microfollicular arrangement and equivocal nuclear features
    • Percentage of tumor to be involved by this pattern is still debated
    • Subcategorized into unencapsulated, encapsulated/well-demarcated and diffuse/multinodular variants
    • Encapsulated—akin to follicular neoplasms, better prognosis
    • Diffuse type more aggressive.
 
Papillary Microcarcinoma
It is not a specific variant but includes all papillary carcinomas that measure one cm or less in dimension (stage IA).
 
Clinical Markers for Potential Aggressiveness
  • Nonincidental presentation
  • Positive preoperative FNA
  • Lymph node metastasis
  • Positive family history
  • Nodules in contralateral lobes
  • Male gender.
 
Histological Markers for Aggressiveness
  • Multifocal/bilateral
  • Size ≥6 mm
  • Extrathyroidal extension
  • Desmoplastic fibrosis
  • Presence of poorly differentiated components
  • Lymphovascular emboli.
7
 
Prognosis
Prognosis of papillary carcinoma is excellent.10 year survival rate is over 90% and for young patients, over 98%. Tall cell and columnar cell variants have a less favorable prognosis than conventional papillary carcinomas.
 
Follicular Carcinoma (Figs 1.3 and 1.4)
It is a malignant epithelial tumor showing follicular cell differentiation and lacking diagnostic nuclear features of papillary carcinoma. It accounts for 10–15% of thyroid malignancies. It is more common in women in the fifth decade. Incidence is higher in iodine deficient areas. Follicular carcinomas most commonly present as large asymptomatic thyroid nodules which are typically cold on scintigraphy. Distant metastasis is seen in up to 20% during presentation. Oncocytic variants typically occur ten years later than the conventional types and show greater propensity for recurrence and local invasion. Follicular carcinomas are usually encapsulated with grey tan to brown bulging cut surface. Widely invasive carcinomas may show extensive permeation of the capsule.
zoom view
Fig. 1.3: Follicular carcinoma (H & E, 10X)
Rarely thyroid veins and superior vena cava may be involved. Multifocality is uncommon. Distal metastasis to the lung and bones are common.
 
Cytology
Aspirates will be hypercellular and show repetitive microfollicles and scant colloid. Atypical nuclear features do not denote malignancy. Demonstration of capsular or vascular invasion is needed for the diagnosis of follicular carcinoma.
 
Histology
Follicular carcinomas show variable morphology with cells arranged in follicles, solid or trabecular patterns. They are divided into two major categories—minimally invasive and widely invasive. While conceptually simple, there is no consensus as to the definition of capsular invasion. Some authorities require complete transgression of the capsule, while other authorities do not require complete transgression of the capsule. Minimally invasive carcinomas have limited capsular and/or vascular invasion. Widely invasive carcinomas have widespread invasion of thyroid tissue and/or blood vessels. The probability of aggressive behavior increases with the extent of vascular invasion.
zoom view
Fig. 1.4: Follicular carcinoma with cells in closely packed microfollicular pattern (H & E, 40X)
The term ‘grossly encapsulated 8angioinvasive follicular carcinoma’ has been suggested for those tumors that demonstrate vascular invasion only.
 
Variants
  • Oncocytic
  • Clear cell
  • Mucinous.
 
Prognosis
Minimally invasive follicular carcinomas have very low long-term mortality (3–5%). Widely invasive carcinomas have 50% mortality. Oncocytic carcinoma behaves more aggressively than conventional types with higher frequencies of extrathyroidal extension, local recurrence and nodal metastasis. Adverse prognostic factors include age >45 years, oncocytic tumor type, extrathyroidal extension, tumor size >4 cm and presence of distant metastasis.
 
Poorly Differentiated Carcinoma
They are defined as follicular cell neoplasms that show limited evidence of follicular cell differentiation and occupy both morphologically and behaviorally an intermediate position between differentiated and undifferentiated carcinomas. Turin proposal (2006) remains so far the most accepted criteria for diagnosing this entity. According to this proposal PDTCs are defined by: (1) Presence of TIS (trabecular/insular/solid) architecture, with (2) At least one of the following features—convoluted nuclei, mitotic figures of >3/hpf, or coagulative necrosis; (3) Absence of conventional nuclear features of papillary carcinoma. PTDCs can be seen as component of well-differentiated carcinomas and as little as 10% is sufficient to confer an aggressive biological behavior. Most tumors present as cold nodules with or without enlarged lymphadenopathy. Lung and bone metastases are also relatively frequent at the time of diagnosis. Extrathyroidal extension is less commonly seen than in anaplastic carcinomas.
Focal TP53 positivity, increased ki67 index (10–30%) and absence of E-cadherin membrane expression are important immunohistochemical features. They show reactivity for both thyroglobulin and TTF1, although thyroglobulin positivity may be focal.
These patients respond poorly to radioiodine therapy. The prognosis depends primarily on TNM staging, completeness of surgery and response to radioactive iodine therapy.
 
Anaplastic (Undifferentiated) Carcinoma (Fig. 1.5)
Anaplastic carcinomas are highly malignant tumors that histologically appear wholly or partially composed of undifferentiated cells that exhibit immunohistochemical or ultrastructural features indicative of epithelial differentiation. It affects mainly the elderly age group with higher incidence reported in endemic goiter regions. It has a high mortality rate (90%) with a median survival rate of up to 6 months after diagnosis.
zoom view
Fig. 1.5: Anaplastic thyroid carcinoma showing bizarre cells, hematoxylin and eosin, 40x
Patients typically present with rapidly expanding neck mass 9with pressure symptoms like hoarseness and dysphagia. Tumors are hard and fixed and frequently invade the surrounding structures. Lymph node involvement as well as distant metastasis is seen in up to 40% of cases. All anaplastic carcinomas are staged as T4 (T4a-intrathyroidal, T4b—extrathyroidal extension).
Cytologically, smears are cellular with highly pleomorphic cells seen singly and in clusters. Three types of cells are observed—spindle, giant cell and squamoid. There is increased mitosis and necrosis with the background characteristically showing polymorphonuclear leukocytes. There may be an associated differentiated component in some of the cases. Variants described are: (1) osteoclastic, (2) carcinosarcoma, (3) paucicellular, (4) lymphoepithelioma like.
These tumors are negative for thyroglobulin and TTF1 and show variable positivity with epithelial markers. Immunohistochemistry is used to differentiate them from other mesenchymal tumors, melanomas and lymphomas.
Prognostic factors are related primarily to the extent of disease at presentation. Prognosis depends on the size of the undifferentiated component and the efficacy of eradicative surgery. 5 year survival rate ranges from 0 – 14%.
 
Medullary Carcinoma (Figs 1.6 to 1.8)
Medullary thyroid carcinoma is a malignant tumor showing C cell differentiation. It constitutes 5–10% of all thyroid malignancies. Up to 25% cases are heritable, caused by germ-line mutations in RET proto-oncogene. Mean age at presentation is 50 years for sporadic cases. MEN II B patients present in infancy or early childhood while MEN IIA associated tumors occur in late adolescence or early adulthood. Patients with FMTC present at an age of 50 years.
zoom view
Fig. 1.6: Medullary carcinoma with cells in nesting pattern (H & E, 10X)
zoom view
Fig. 1.7: Salt and pepper chromatin in medullary carcinoma (H & E, 10X)
zoom view
Fig. 1.8: Amyloid deposits in medullary carcinoma (H & E, 40X)
Tumors typically 10are located in the middle third of the lobes. They present as cold nodules with more patients presenting with nodal metastasis (50%) and up to 15% with distant metastasis. Virtually all MTCs produce calcitonin and serum levels are typically increased. Paraneoplastic syndromes may occur due to production of other peptides and amines. Cytologically smears show loosely cohesive cells with polygonal, bipolar, or spindle shapes. Plasmacytoid cells are common and multinucleated giant cell morphology is occassionally encountered. MGG stains show characteristic red cytoplasmic granules. Amyloid may be found in 50–70% of the cases.
Histologically, cells with salt and pepper chromatin are arranged in sheets, nests or trabeculae in an organoid fashion. Necrosis is infrequent. Variants include spindle cell, small cell, giant cell, oncocytic, clear cell, etc. (12 variants are described). Occasional cases may show more pleomorphic features.
Cells are positive for calcitonin, CEA, chromogranin, synaptophysin, TTF1 and low molecular weight keratin. C cell hyperplasia can be seen in surrounding thyroid tissue adjacent to the invasive tumor and also in prophylactic thyroidectomies in hereditary cases. ‘Neoplastic C cell hyperplasia is a precursor lesion for heritable medullary carcinomas and is composed of groups of intrafollicular atypical C cells while ‘reactive C cell hyperplasias’ seen in variety of other pathophysiological conditions is characterized by an increased number of normal appearing C cells.
Five and 10 year survival rates are 83.2% and 73.7%, respectively. Older age, male gender and extent of local tumor invasion are associated with reduced survival. Presence of distant metastasis is also an independent predictor of poor prognosis. Children with MEN IIB present at an earlier age and have a higher risk for aggressive forms when compared with MEN IIA cases. Presence of necrosis, squamous metaplasia, <50% of calcitonin reactive cells or CEA reactive cells in the absence of calcitonin has also been considered as a poor prognostic feature.
 
Molecular Genetics
Alterations of follicular cells that lead to carcinogenesis are caused by unopposed activation of either the mitogen-activated protein (MAP) kinase pathway or the phosphatidylinositol-3-kinase (PI3K)/AKT pathway. Specifically, the MAP kinase pathway (encompassed by the MEK and ERK kinase cascade) is regulated by the RET, RAS, and BRAF genes. Point mutations in the BRAF and RAS genes or RET/PTC translocation can lead to unopposed cellular proliferation and to a carcinogenic environment via the MAP kinase pathway.
BRAF gene alterations are present in about 30% to 45% of patients with PTCs and, on average, in 15% (range, 12%–47%) of patients with PDTCs. The BRAF gene is a marker of adverse prognostic factors, including disease aggressiveness, decreased radioiodine trapping, tumor recurrence, lymph node or distant metastatic disease, and extrathyroidal extension. RET/PTC rearrangements seen in 30% of PTC ultimately result in the unopposed activation of the MAP kinase pathway.
PAX8: PPARγ rearrangements are almost always associated only with follicular carcinomas. Such rearrangements are almost never expressed in patients with PDTCs.
RAS gene alterations are present in about 40% to 50% of patients with FTCs and, on average, in 35% (range, 20–50%) of patients with PDTCs and ATCs. It is a marker of tumor dedifferentiation and adverse prognostic outcome. TP53 gene alterations are rarely associated with WDTCs; however, they are highly prevalent in patients with PDTCs (about 28%; range, 17–38%) and in patients with ATCs (64%; range, 20–88%). Unlike the RAS and BRAF gene alterations, p53 mutations possess an exclusive function in triggering tumor 11dedifferentiation and evolution to PDTC and ATC.
Table 1.1   Summary of the prevalence of genetic alterations in patients with various thyroid carcinomas
Altered gene
PDTC
PTC
FTC
ATC
RET/PTC
0%
20%
0%
0%
TP53
20–30%
0%
0%
65–70%
BRAF
15%
45%
0%
20–25%
RAS
30–35%
10–15%
45%
50–55%
ß-catenin
20–25%
0%
0%
65%
PAX8: PPARγ
0%
0%
35%
0%
The molecular genetics of medullary carcinoma is well-established, showing mutations in the RET proto-oncogene. Germline RET mutations are associated with hereditary medullary carcinomas, including familial medullary carcinoma (familial MTC) and the multiple endocrine neoplasia syndromes (MEN2a and 2b). However, it must also be noted that sporadic tumors may also harbor RET mutations (30–66%). Sporadic tumors may also harbor HRAS or KRAS mutations as well (up to 25%).
In familial setting, prophylactic total thyroidectomy is performed for family members based on positive mutational analysis.
Table 1.1 summarizes the prevalence of genetic alteration in patients with various thyroid carcinomas.
 
Ancillary Testing
Ancillary testing can be used for diagnostic, prognostic, and, to some extent, therapeutic purposes in thyroid cancer. While several markers are now commonly used in patient management, they are not yet a “universal standard of care.”
A number of immunohistochemical markers have been proposed to confirm the diagnosis of papillary carcinoma, allowing for distinction from other lesions/tumors in the differential diagnosis. These markers include HBME-1, galectin 3, CITED-1, HMWCK, CD56 and cytokeratin 19. The literature has demonstrated a high sensitivity and specificity with various combinations of these markers for the diagnosis of papillary carcinoma and is particularly useful in resolving the diagnosis for follicular patterned lesions. However, these panels are not infallible as there are false-positives and false-negatives. With regard to the rare but important cribriform morular variant of papillary carcinoma, nuclear beta catenin accumulation is essentially a defining feature that is diagnostically invaluable.
Further Reading
  1. Demellawy DE, Nasr A, Alowami S. Application of CD56, P63 and CK19 immunohistochemistry in the diagnosis of papillary carcinoma of the thyroid. Diagnostic Pathology, 2008.
  1. CAP guidelines for thyroid cancer reporting Aug 2014.
  1. Legakis I, Syrigos K. Recent Advances in Molecular Diagnosis of Thyroid Cancer. J Thyroid Res. vol. 2011, Article ID 384213, 8 pages, 2011. doi:10.4061/2011/384213.
  1. Hannallah J, Rose J, Guerrero MA. Comprehensive Literature Review: Recent Advances in Diagnosing and Managing Patients with Poorly Differentiated Thyroid Carcinoma. Internat J Endocrinol. vol. 2013, Article ID 317487, 7 pages, 2013. doi:10.1155/2013/31748.
  1. Papillary thyroid carcinoma: An update, Virginia A LiVolsi Modern Pathology. 2011;24:S1-S9.
  1. RC Path guidelines for thyroid cancer reporting; 2014.

  1. 12 Fischer S, Asa SL. Application of Immunohistochemistry to Thyroid Neoplasms. Arch Pathol Lab Med. 2008;132:359–72.
  1. The Bethesda System for Reporting Thyroid Fine-Needle Aspiration Specimens AJCP, 2010 134:343–344; doi:10.1309/AJCPXM9WIRQ8JZBJ.
  1. WHO bluebooks-Endocrine organs, 3rd edition -2004.
  1. Xing M, Haugen BR, Schlumberger M. Progress in molecular-based management of differentiated thyroid cancer. Lancet. 2013;381(9871):1058–69. doi:10.1016/S0140–6736(13)60109–9.