Cancer of the Endocrine System


Summary of Key Points

  • Thyroid Cancer

  • The incidence of thyroid cancer is increasing, and there are approximately 33,500 new cases per year in the United States.

  • The incidence of differentiated thyroid cancer is 14.3 per 100,000 people per year, with a female-to-male ratio of more than 3 : 1.

  • Differentiated thyroid cancer (DTC) includes papillary thyroid cancer (PTC), which accounts for 80% of all thyroid cancers; follicular thyroid cancer (FTC), which accounts for 10% to 20% of all thyroid cancers; and a rare type, Hürthle cell cancer.

  • Medullary thyroid cancer (MTC) arises from the parafollicular C cells and accounts for 1% to 2% of all thyroid cancers.

  • Anaplastic thyroid cancer is a rare, but rapidly fatal, form of thyroid cancer.

  • Other histologic types of cancer, such as lymphoma, sarcoma, and metastatic cancers, can also be found within the thyroid.

  • Known risk factors for the development of thyroid cancer include radiation exposure and iodine deficiency.

  • Thyroid cancer can also run in families or exist as part of familial syndromes (Gardner, Cowden, and Werner syndromes).

  • More recently, the molecular pathogenesis of thyroid cancer has been investigated. The following are the most widely studied molecular markers for DTC to date:

    • RET/PTC rearrangement

    • BRAF

    • PAX8-PPARγ rearrangement

    • NRAS, KRAS and HRAS

  • Cancer with well-differentiated histologic features has an excellent 5-year survival rate (>95%).

  • Older age and extent of invasion are related to prognosis.

  • Lymph node involvement is associated with higher recurrence but has questionable impact on survival.

  • Multiple staging systems exist for DTC.

  • For Hürthle cell adenoma, larger size (>6 cm) is predictive of malignancy.

  • For poorly differentiated tumors, lymph node metastases are common in recurrence.

  • Anaplastic cancers are extremely aggressive, with 5-year survival rates below 5%.

  • The history should include radiation exposure, family history, and compressive symptoms (dysphagia, hoarseness, pain or pressure) from enlarging tumor.

  • Concerning examination findings include a fixed mass or lymphadenopathy.

  • Presented as incidental findings on computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET), and ultrasound.

  • Preoperative laboratory studies include thyroid-stimulating hormone (TSH) and thyroglobulin (Tg).

  • Fine-needle aspiration (FNA) biopsy is a key component of the workup of thyroid nodules. The Bethesda criteria classify FNA results and determine the risk of cancer in the nodule.

  • Preoperative imaging should include cervical ultrasound. CT is used when aggressive variants are suspected, in order to assist in operative planning.

  • Treatment begins with surgery. Most thyroid cancers are treated with total thyroidectomy. Compartment-oriented neck dissection is added when there is metastatic disease in the cervical lymph nodes.

  • Adjuvant therapy for differentiated tumors is radioactive iodine (iodine-131 [ 131 I]).

  • Patients must be prepared for radioactive iodine ablation with low iodine diet, withdrawal from thyroid hormone replacement, or given recombinant human thyroid hormone (rhTSH) if there is no evidence of metastatic disease.

  • After surgery and radioactive iodine, thyroxine suppression prevents the growth of microscopic disease.

  • External beam radiation is used for persistent, recurrent, anaplastic, poorly differentiated tumors that are not iodine avid.

  • Chemotherapy is mainly palliative for poorly differentiated or anaplastic tumors. Traditional chemotherapy has minimal response rates, but newer, targeted therapies, such as sorafenib, lenvatinib, and sunitinib, are showing promise.

  • Surveillance for recurrent thyroid cancer includes measurements of TSH, Tg, and anti-Tg antibodies in addition to cervical ultrasound. The schedule of these tests is tailored to risk level.

  • Treatment of recurrence can include external beam radiation, targeted therapies, or ultrasound-guided laser ablation, depending on the iodine avidity of the tumor.

  • Medullary Thyroid Cancer

  • Medullary thyroid cancer (MTC) accounts for 1% to 2% of all thyroid cancers; 75% of cases are sporadic, and 25% are familial (multiple endocrine neoplasia [MEN] type 2, familial medullary thyroid carcinoma [FMTC]).

  • The diagnosis is made by FNA with calcitonin staining and/or washout. RET testing can identify inherited germline mutations. Cervical ultrasound or CT scans assist with operative planning. The tumor markers calcitonin and carcinoembryonic antigen (CEA) can be useful in following patients postoperatively for identifying recurrence and metastases.

  • At a minimum, treatment of clinical MTC should consist of total thyroidectomy plus central lymph node dissection. Lateral neck dissection is added when there are clinically positive nodes in the central neck and for high-risk patients.

  • Traditional chemotherapy is not effective for metastatic MTC, but newer, targeted therapies for metastatic disease, such as vandetanib and cabozantinib, have shown some promise.

  • Adrenocortical Cancer

  • The incidence of adrenocortical cancer is 1 to 2 per million people.

  • Most adrenocortical cancers are sporadic, but they can also occur as part of familial syndromes such as MEN1, Li-Fraumeni syndrome, Beckwith-Wiedemann syndrome, and Carney complex.

  • Most are asymptomatic, but 40% to 60% are functional (hormone production), and this may be the presenting symptom(s).

  • The diagnosis is by urinary or plasma biochemical testing and imaging: CT, fluorodeoxyglucose (FDG)-PET.

  • Often the diagnosis is not made definitively until after resection of suspicious masses, and pathologic assessment provides definitive diagnosis. FNA of adrenal masses is rarely indicated.

  • Surgery is the mainstay of treatment for adrenocortical cancer and should consist of en bloc resection of the adrenal gland with adjacent organs and tissue that is involved; cardiopulmonary bypass may be necessary for caval involvement.

  • Long-term surveillance, consisting of physical examinations and CT scans, is necessary to monitor for disease recurrence.

  • Mitotane alone or in combination with other chemotherapeutic agents improves recurrence-free survival.

  • Radiotherapy may improve local control, but there are no clear recommendations.

  • Hormonal control can also limit disease spread and consists of mitotane, ketoconazole, metyrapone, and etomidate.

  • Excision or reoperation is recommended for recurrent or metastatic disease.

  • The prognosis is poor, with an overall 5-year survival rate of less than 40%.

  • Malignant Pheochromocytoma

  • The incidence of malignant pheochromocytoma (PCC) is 2 to 8 per 1,000,000 adults.

  • Most malignant PCCs are sporadic, but 10% are part of inherited syndromes such as MEN syndromes, neurofibromatosis type 1, von Hippel-Lindau syndrome, and succinate dehydrogenase gene mutations.

  • PCCs manifest with the classic triad of functional tumors—headache, tachycardia, and sweating—but they are asymptomatic in more than 50% of cases, incidentally discovered as an adrenal mass.

  • The diagnosis is established with urinary or plasma fractionated metanephrines and catecholamines.

  • Tumors are localized with CT or MRI; metaiodobenzylguanidine (MIBG) is used to identify extraadrenal metastases.

  • Treatment begins with surgery to resect the entire gland with clear margins. Surgery can also be useful in debulking for metastatic disease.

  • Metastatic or unresectable disease can be treated with 131 I-MIBG or chemotherapy.

  • Radiotherapy is used for palliation in bone and lymph node metastases.

  • Medical therapy to prepare patients for surgery or control symptoms of catecholamine excess can include phenoxybenzamine, nicardipine, or metyrosine.

  • Five-year survival ranges from 20% to 50%.

  • Multiple Endocrine Neoplasia Syndromes

  • MEN1 manifests first with hyperparathyroidism in patients in their 30s and 40s. Other manifestations include pituitary tumors and neuroendocrine tumors (NETs) of the pancreas (such as gastrinoma) and upper gastrointestinal tract.

  • MEN1 is inherited in an autosomal dominant fashion. Mutations in the MENIN tumor suppressor gene cause this disease, but expression is variable.

  • Treatment of parathyroid hyperplasia is subtotal parathyroidectomy and bilateral cervical thymectomy.

  • MEN1 patients with pancreatic and duodenal tumors are treated with distal pancreatectomy, enucleation of pancreatic head tumors, duodenotomy, and mucosal resection of multiple duodenal tumors.

  • MEN2A is characterized by PCC, MTC, and primary hyperparathyroidism (hyperplasia).

  • MEN2B is characterized by more aggressive MTC, PCC, and mucosal ganglioneuromas.

  • The MTC in MEN2 syndromes arises from C-cell hyperplasia and germline RET mutations.

  • Specific codon mutations in the RET gene determine the disease phenotype in MEN2 syndromes, and help risk-stratify patients.

  • Prophylactic thyroidectomy should be offered to mutation carriers; the timing of thyroidectomy is determined by the specific codon mutation.

  • PCCs are often bilateral in MEN2 syndromes, but onset is asynchronous. Consequently, prophylactic adrenalectomy is not indicated. Those MEN2 patients who develop bilateral disease can be treated with bilateral adrenalectomy and hormone replacement or cortical-sparing adrenalectomy.

  • Carcinoid Tumors

  • The incidence of carcinoid tumors is 5.25 per 100,000 people.

  • Tumors are identified with specific immunohistochemical staining for neuron-specific enolase (NSE) or chromogranin A. Chromogranin A also serves as a blood marker for the disease.

  • Several classification systems exist for carcinoid tumors, including the World Health Organization (WHO) classification and the European Neuroendocrine Tumor Society (ENETS) staging system.

  • Carcinoids arise from the Kulchitsky cells in crypts of Lieberkühn of the gut or disseminated in the endobronchial mucosa, and are classified by location.

  • The diagnosis is made definitively through tissue diagnosis, but urinary 5-hydroxyindoleacetic acid (5-HIAA), serum NSE, and chromogranin A are serum markers of the disease.

  • CT or MRI can be used to localize the carcinoid tumors, gallium-68 ( 68 Ga)–DOTATATE PET-CT or OctreoScan can be used, because these tumors have somatostatin receptors.

  • The carcinoid syndrome occurs in metastatic carcinoid and presents as flushing, diarrhea, and bronchoconstriction. Right-sided valvular heart disease is also a manifestation of the disease.

  • Treatment begins with resection of primary tumor with nodal metastases.

  • Debulking or metastasectomy is beneficial for controlling symptoms in patients with liver disease or bulky disease.

  • Radiation therapy is rarely used for primary therapy but can be palliative.

  • Antihormonal therapy consists of octapeptide analogues of somatostatin; Sandostatin LAR is a helpful, long-acting formulation octapeptide analogue of somatostatin.

  • The liver is a common site for metastatic carcinoid, and there are several options for hepatic-directed therapy, including surgery and embolization (chemoembolization or radioembolization).

  • Metastatic disease can also be treated with targeted agents or emerging radionuclide therapy.

  • Pancreatic Neuroendocrine Tumors

  • The incidence of NETs is 2.4% to 5.8% per 100,000 people.

  • Pancreatic neuroendocrine tumors (pNETs) can be sporadic or inherited (MEN).

  • pNETs are diagnosed with CT or MRI imaging; ultrasound or endoscopic ultrasound (EUS) examination can help guide biopsy.

  • The ENETS staging system is proposed to help stage pNETs.

  • Insulinomas are diagnosed with fasting hypoglycemia with elevated plasma insulin levels; 10% are malignant, and surgical resection (enucleation) is curative.

  • Glucagonoma is characterized by migratory necrotizing erythema, insulin-resistant diabetes, glossitis, ileus, and constipation; 50% to 80% are metastatic.

  • Because of the higher rate of metastatic disease, surgical resection is curative in less than one-third of patients with glucagonoma.

  • Somatostatinoma is characterized by diabetes, diarrhea, and gallbladder disorders.

  • Treatment for somatostatinoma includes cytoreductive surgery and chemotherapy.

  • Gastrinoma is characterized by ulcer disease, in spite of adequate treatment, and diarrhea.

  • Gastrinoma is diagnosed through hypergastrinemia with elevated basal acid output or positive secretin test result; tumors are localized with CT, MRI, or octreotide scan. These tumors are frequently metastatic.

  • Targeted therapies such as everolimus and sunitinib have been approved by the US Food and Drug Administration (FDA) for first-line treatment of pNETs.

  • Parathyroid Carcinoma

  • The incidence of parathyroid carcinoma is 5.73 per 10 million people.

  • The etiology of parathyroid cancer has recently been attributed to pericentromeric inversion resulting in overexpression of the cyclin D1 gene.

  • In hyperparathyroidism–jaw tumor syndrome, there are mutations in the HRPT2 tumor suppressor gene (parafibromin).

  • Clinical characteristics of parathyroid carcinoma include the constitutional symptoms of primary hyperparathyroidism, including muscle weakness, fatigue, nausea, vomiting, increased thirst, and frequent urination, in addition to bone pain and fractures.

  • A neck mass occurs in 34% to 52% but is uncommon in benign parathyroid adenomas.

  • In parathyroid carcinoma, serum calcium is quite elevated (14.6–15.9) with elevated serum parathyroid hormone (PTH) (commonly 10-fold higher than the upper limit of normal).

  • The diagnosis is made by means of laboratory measurement of Ca and PTH, and then technetium-99m sestamibi scan and neck ultrasound can be used to localize the tumor with or without washout for PTH measurement in FNA material.

  • Pathologic features of parathyroid carcinoma include local invasion and lymph node metastases.

  • Treatment of parathyroid carcinoma should include en bloc resection of the parathyroid mass with ipsilateral thyroid lobe with or without ipsilateral neck dissection followed by postoperative calcium and activated vitamin D supplementation.

  • Medical therapy for hypercalcemia precipitated by parathyroid carcinoma should start with hydration and loop diuretics; calcimimetics (Cinacalcet) or bisphosphonates can later be added to lower the serum calcium levels.

  • Adjuvant therapy includes chemotherapy, such as dacarbazine, 5-fluorouracil (5-FU), or cyclophosphamide; radiotherapy is of limited efficacy.

  • Patients with features of hyperparathyroidism–jaw tumor syndrome or a family history should undergo genetic counseling and HRPT2 testing.

  • After surgery, one-third of patients are cured, one-third have recurrence after prolonged disease-free survival, and one-third experience a short, aggressive course; the 5-year survival is 82.5%.

Unlike tumors found elsewhere in the body, cancers of the endocrine organs can cause symptoms of hormonal excess in addition to mass effect, obstruction, or pain from the mass itself. Therefore physicians who care for patients with endocrine cancers must combat both the physiologic manifestations and the neoplasia. Any of the endocrine tumors can be part of a multitude of familial syndromes, so the clinician caring for these patients must always keep this possibility in mind.

This chapter covers selected endocrine cancers and highlights some of the unique challenges in treating such tumors. Many common themes emerge. Diagnosis typically involves biochemical confirmation of the endocrinopathy followed by imaging to locate the tumor(s). Surgery plays a role in the initial treatment and often for recurrence and palliation. Traditional chemotherapy is of limited use for endocrine cancers, but newer, targeted therapies show more promise. This chapter covers thyroid cancer, including medullary thyroid cancer (MTC), adrenocortical carcinoma, malignant pheochromocytoma (PCC), multiple endocrine neoplasia (MEN) syndromes, carcinoid tumors, pancreatic neuroendocrine tumors (pNETs), and parathyroid carcinoma.

Thyroid Cancer

Incidence

Thyroid cancer is the most common endocrine cancer. A spectrum of biologic behavior exists, ranging from indolent, well-differentiated tumors to extremely aggressive, poorly differentiated or anaplastic cancers. Thyroid cancer is the most rapidly increasing malignancy in the United States for both men and women. From 1980 to 2006, the annual US thyroid cancer age-adjusted incidence rose from 4.33 to 11.03 cases per 100,000 population. This incidence increased by 2009 to 14.3 per 100,000. This is a nearly threefold increase in incidence. The gender-adjusted incidence rose from 6.5 to 21.4 = 14.9 per 100,000 women and almost 4 times greater than that of men, from 3.1 to 6.9 = 3.8 per 100,000 men. This increasing incidence is attributed to improved detection of smaller tumors, mostly papillary thyroid cancer, using high-resolution neck ultrasonography. This change has been attributed mostly to the increase of papillary thyroid cancer (PTC) incidence. A published report from a population-based study states that the rapid increase in incidence of thyroid cancer is attributed to occult cancer detected through neck imaging with stable clinically detected thyroid cancer and disease mortality. Despite this improved detection, the mortality rate remains unchanged at 0.5 per 100,000 population. Therefore, thyroid cancer presents a unique challenge to the treating physician to manage patient expectations, minimize potentially lifelong complications, use the appropriate surveillance for follow-up, and identify patients with more aggressive, poorly differentiated forms.

Classification

The most common type of thyroid cancer is PTC, representing 80% of all cases. The second most common type is follicular thyroid cancer (FTC), which represents 10% to 20% of all cases. Together, papillary and follicular cancers are termed differentiated thyroid cancer (DTC), and both arise from the thyroid follicular cells. MTC comes from the parafollicular C cells. This neuroendocrine thyroid tumor represents 5% to 10% of all thyroid cancer cases and occurs in familial and sporadic forms. Finally, anaplastic thyroid cancer (ATC) is one of the most aggressive and rapidly fatal cancers. It can develop from DTC that dedifferentiates over time, and it also arises de novo. The first part of this section on thyroid cancer discusses DTC, and the second part reviews MTC ( Table 68.1 , Fig. 68.1 ).

Table 68.1
Histologic Classification of Thyroid Cancers and Their Incidence
Tumor Histology Incidence (%)
Differentiated carcinomas 81–87
Papillary
Follicular variant of papillary
Follicular and Hürthle cell
Medullary 6–8
Anaplastic 5
Lymphoma 1–5
Metastatic <1

Figure 68.1, Histologic patterns of thyroid cancer. (A) Papillary carcinoma. (B) Pure follicular carcinoma. (C) Anaplastic carcinoma. (D) Medullary carcinoma.

Etiology

External radiation exposure to the cervical region is one of the most well-known causes of thyroid cancer. This risk is related to radiation dose and age, and persists throughout life. Irradiation could result from an external source used for diagnostic and therapeutic purposes or from internal radiation from food or liquid that has radioactivity. Historically, patients received radiation treatments for enlarged tonsils or facial acne. Today, patients with cancer such as Hodgkin disease might still receive radiation treatments. In addition, children exposed to radioactive fallout from the Chernobyl (Russia) accident have demonstrated an increased incidence of thyroid cancer. Based on evidence from patients radiated for Hodgkin disease, doses of 40 Gy are potentially carcinogenic. Epidemiologic studies have reported that 7% to 9% of patients who received 5 to 10 Gy of external beam radiation develop thyroid cancer. A lag time of 10 to 20 years usually exists between exposure and diagnosis of thyroid cancer, although much shorter periods have been reported ( Table 68.2 ).

Table 68.2
Risk Factors for Malignancy in Nodular Thyroid
Modified from Sessions RB, Diehi WL. Thyroid cancer and related nodularity. In: Myers E, Suen J, es. Cancer of the Head and Neck. 2nd ed. New York: Churchill Livingstone; 1981:766.
Factor L ow R isk H igh R isk
1 2 3 4 5
Age
Elderly
Child
Sex
Male
Female
Low-dose radiation in childhood
Family history
Cystic mass
Solid mass
Multiple masses
Solitary mass
Growing mass
Stable mass
Hot scan
Cold scan
Warm scan
Fine-needle aspiration (−)
Fine-needle aspiration (+)
Associated cervical adenopathy
Complete resolution in response to thyroid suppression
Partial resolution in response to thyroid suppression
No response to suppression

Included in the environmental etiology for thyroid cancer is dietary iodine content. A higher incidence of PTC exists in regions with high dietary iodine content, such as the Pacific rim and Iceland. Iodine-deficient countries, in contrast, experience a higher incidence of FTC in addition to benign thyroid goiters. Many factors confound these studies that link changes in DTC rates to iodine intake. Ethnicity, selenium, goitrogen, and carcinogen intake likely play causative roles.

Increasing investigation into molecular markers that can distinguish carcinoma from benign nodules has led to a greater understanding of the genetic alterations in thyroid cancer. For example, 70% of cancers found in Chernobyl survivors carried an RET and PTC gene (RET/PTC) rearrangement. The fusion of the tyrosine kinase encoding domain of the RET protein with a heterologous group of genes occurs in 20% to 40% cases of PTC and is called the RET/PTC rearrangement. RET/PTC rearrangements are frequent in small, multifocal PTCs accompanied by an inflammatory infiltrate, often seen in individuals exposed to ionizing radiation and in children. BRAF is a member of the RAF-MEK-ERK serine/threonine kinase-signaling cascade, and a BRAF mutation is found in 40% to 60% of PTC cases. The V600E BRAF point mutation or mutations in another member of this signaling pathway, RAS, are frequent in cases of poorly differentiated PTC or ATC. Analysis of 27 PTCs by Nikiforova and colleagues showed that 70% harbored mutated genes, distributed among BRAF (60%), PIK3A (11%), TP53 (7%), and NRAS (4%). In the same study, the prevalence of mutated genes in follicular carcinoma was NRAS, 2 5%; KRAS, 0.5%; HRAS, 0.2%; TSHR, 11%; TP53, 11%; and PTEN, 0.2%. Most BRAF substitutions keep the protein in a catalytically active form, resulting in constitutive activation of the RAF-MEK-ERK signaling cascade and constant mitogenic activity. A very high specificity of approximately 99% of BRAF V600E has been estimated in data from multiple studies, with very low sensitivity to rule out malignancy.

A higher prevalence of BRAF V600E mutations were observed in papillary thyroid microcarcinoma with lymph node metastasis and tumor recurrence.

The Ras proteins are plasma membrane guanosine triphosphatases activated by growth factor receptors. Mutations that result in their constitutive activation lead to oncogenesis. RAS mutations occur in approximately 40% of follicular cancers and in a small portion of PTCs, particularly the follicular variant of PTC. Similar to the RET/PTC rearrangement, another interchromosomal translocation occurs in FTC. The promoter element of the gene encoding paired box 8 (PAX8) fuses with the coding sequence of the peroxisome proliferator-activated receptor γ (PPARγ) gene in 35% of FTCs. The functional consequences of the PAX8/PPARγ rearrangement remain unclear. RAS mutations are also highly prevalent in FTC, but Nikiforova and colleagues found that RAS and PAX8/PPARγ rearrangements are mutually exclusive, suggesting that these are two distinct molecular pathways for FTC development.

For diagnostic accuracy of molecular markers, especially for indeterminate thyroid nodules, multiple panels have been expanded to include mutational/translocational BRAF, NRAS, HRAS, and KRAS point mutations, in addition to RET/PTC1 and RET/PTC3, with or without PAX8/PPARγ rearrangements. TERT promoter mutations have been identified in 2% to 10% of sporadic PTCs. They may coexist with the V600E BRAF mutation in PTC and tend to be more aggressive.

Aside from these acquired genetic lesions, some forms of DTC are also inherited. DTC is seen in familial syndromes such as Gardner syndrome, Cowden syndrome, and Werner syndrome. Familial nonmedullary thyroid cancer (FNMTC), in which two or more first-degree relatives have been diagnosed with DTC in the absence of another syndrome, exhibits autosomal dominant behavior with incomplete penetrance and variable expressivity. Linkage analyses have identified several candidate genes for FNMTC, including TCO1, MNG1, fPTC/PRN, and NMTC1, but a single responsible gene has not been identified. Evidence has shown that 5% to 10% of DTCs have familiar occurrence. Compared with sporadic DTC, FNMTC is more aggressive with increased recurrence and decreased disease-free survival, local invasion, multicentricity, lymph node metastases, invasion to surrounding structures, and combination with chronic lymphocytic thyroiditis. However, in the absence of a suitable genetic test, families cannot be screened, and FNMTC is difficult to distinguish from sporadic DTC.

Classification and Prognosis

DTCs are divided broadly as papillary or follicular. Follicular variant PTC has features of both PTC and FTC but is classified as an PTC subtype (see Table 68.1 and Fig. 68.1A ). In general, well-differentiated PTC has an excellent prognosis, with 5-year survival greater than 97%. Smaller tumors carry a better prognosis than larger tumors. PTCs smaller than 1 cm are called papillary microcarcinomas and have been reported in 10% to 30% of autopsy studies. In the past, these tumors were incidentally detected in thyroidectomy specimens, but they are now detected with increasing frequency with high-resolution ultrasonography. They are believed to have an excellent prognosis, with a structural disease recurrence rate of 1% to 2% in unifocal PTCs and 4% to 6% in multifocal PTCs. However, some may behave more aggressively than previously appreciated, and management remains controversial.

Age is another important determinant of prognosis in DTC. Older patients tend to have more poorly differentiated, aggressive variants and are less likely to respond to radioactive iodine (RAI). In these cases, death results from local invasion and extensive metastases. Therefore the completeness of resection and extrathyroidal extension are two prognostic indicators used in many staging systems for DTC.

The role of lymph node metastases in determining DTC-specific survival remains controversial. Lymph node involvement is common in PTC, but the exact incidence of lymph node metastases depends on how it is defined. Palpable disease in the lymph nodes is present in 5% to 10% of patients with PTC, but ultrasound detects pathologically positive lymph nodes in 30% of patients. Ultrasound features that are associated with thyroid cancer include nodule hypoechogenicity compared with the surrounding structures, microcalcifications, irregular margins, and taller-than-wide shape as measured on a transverse view. Only 2% of patients with FTC have lymph node metastases because the route of spread is mostly hematogenous, but treatment guidelines and retrospective studies frequently consider PTC and FTC together. Routine histologic examination of lymph nodes reveals DTC in 20% to 50% of patients (particularly papillary carcinoma), but when more detailed inspection is performed, up to 90% of patients with DTC will have lymph nodes with microscopic disease, smaller than 2 mm. An increasing amount of high-quality evidence supports sonographic survey of cervical lymph nodes in all patients with thyroid nodules. Historically, lymph node involvement was believed to increase local recurrence without affecting survival, and therefore surgeons took a conservative approach to lymph node dissection for DTC. Wada and colleagues demonstrated that patients with pathologically positive lymph nodes had a recurrence rate of 16.3% compared with 0% in patients without pathologically positive lymph nodes. Whether metastatic lymph nodes are evident preoperatively appears to be an important factor determining recurrence. For example, Ito and colleagues found that if metastatic lymph nodes were not seen preoperatively, then the risk of nodal recurrence was only 1.5%. Of note, in this study of 590 patients with microcarcinomas, 40% of patients had lateral neck lymph node metastases identified histologically after prophylactic neck dissection. Hence, lymph node metastases do affect recurrence, and clinically apparent nodes are more important than pathologically positive nodes. The impact of lymph nodes on survival is less clear. Large series and population-based studies have suggested that there is a small but significant effect on survival.

From the Surveillance, Epidemiology, and End Results (SEER) database, comprehensive analysis showed that patients younger than 45 years with lymph node metastasis had small but significant risk of death compared with younger patients with no lymph node involvement. Because of the questionable effect on mortality, lymph node status is not included in all of the staging systems available for DTC. For example, the AGES system considers age, grade, extrathyroidal extension, and size. The AMES system uses age, distant (non–lymph node) metastases, extent of primary tumor, and size. Some, such as the MACIS system (metastases, age, complete excision, invasion, and size), also account for the adequacy of surgical treatment. Alternatively, staging systems developed by the Ohio State University, the European Organisation for Research and Treatment of Cancer (EORTC), the National Thyroid Cancer Treatment Cooperative Study (NTCTCS), and the American Joint Committee on Cancer (AJCC) all do consider lymph node status. The AJCC staging system is the most widely used ( Table 68.3 ). It is also known as the TNM system because it considers tumor size (T), lymph node metastases (N), and distant metastases (M). Like many of the other thyroid cancer staging systems, it also considers age, with two different classifications for those younger and older than 45 years. In those younger than 45 years, patients with lymph node metastases are classified as stage I unless they have distant metastases (stage II) ( Table 68.4 ).

Table 68.3
TNM Classification of Malignant Tumors of the Thyroid Gland
Modified from Greene FL, Page DL, Fleming ID, et al, eds. AJCC Cancer Staging Manual. 6th ed. New York: Springer-Verlag; 2002.
PRIMARY TUMOR (T STAGE)
Tx Tumor cannot be assessed
T0 No clinical evidence of tumor
T1 Tumor ≤2 cm limited to the thyroid
T2 Tumor >2 cm and <4 cm limited to the thyroid
T3 Tumor ≥4 cm limited to the thyroid or any tumor with minimal extrathyroid extension
T4a Tumor of any size extending beyond the thyroid capsule to invade the subcutaneous soft tissues, larynx, trachea, esophagus, or recurrent laryngeal nerve
T4b Tumor that invades prevertebral fascia or encases carotid artery or mediastinal vessels
ANAPLASTIC CARCINOMAS a
T4a Intrathyroidal—surgically resectable
T4b Extrathyroidal—surgically unresectable
REGIONAL LYMPH NODES (N STAGE)
Nx Regional nodes cannot be assessed
N0 No palpable nodes
N1 Regional nodal metastases
N1a Level VI nodes (pretracheal, paratracheal, prelaryngeal)
N1b Metastasis to unilateral, bilateral, or contralateral cervical or superior mediastinal nodes
DISTANT METASTASES (M STAGE)
Mx Metastases cannot be assessed
M0 No evidence of distant metastases
M1 Distant metastases present

a All anaplastic carcinomas are considered T4 tumors.

Table 68.4
Staging of Thyroid Cancer
From Greene FL, Page DL, Fleming ID, et al, eds. AJCC Cancer Staging Manual. 6th ed. New York: Springer-Verlag; 2002.
Stage TNM
PATIENTS YOUNGER THAN 45 YEARS
I Any T, any N, M0
II Any T, any N, M1
PATIENTS AGE 45 YEARS OR OLDER
I T1, N0, M0
II T2, N0, M0
III T3, N0, M0
T1–3, N1a, M0
IVA T4a, N0–1a, M0
T1–4a, N1b, M0
IVB T4b, any N, M0
IVC Any T, any N, M1
MEDULLARY THYROID CANCER
I T1, N0, M0
II T2–3, N0, M0
III T1–3, N1a, M0
IVA T4a, N0–1a, M0
T1–4a, N1b, M0
IVB T4b, any N, M0
IVC Any T, any N, M1
ANAPLASTIC CANCER
IVA T4a, any N, M0
IVB T4b, any N, M0
IVC Any T, any N, M1

It is worth noting some important ways in which FTC differs from PTC (see Table 68.1 , Fig. 68.1B ). Pure follicular carcinoma tends to occur more in older patients, and it carries a worse prognosis than PTC. Follicular carcinoma is more common in women than in men by approximately three times. Even when the disease is confined to the thyroid, 5% to 15% of patients ultimately die from the disease, although survival still extends decades as in PTC. In addition to the prognostic factors common to the aforementioned DTC staging systems, prognosis in FTC depends on the degree of capsular and vascular invasion. Minimally invasive tumors are grossly contained within the thyroid but have microscopic foci of invasion into the capsule. Invasive tumors carry a worse prognosis and invade the capsule and vessels.

Hürthle cell tumors of the thyroid are often classified with follicular cancer because they are derived from the follicular cell. Both adenomas and carcinomas of the Hürthle cell can occur, and differentiating them by cytologic assessment is difficult, as it is with follicular lesions. Capsular and vascular invasion distinguish carcinoma from adenomas. Large Hürthle cell cancers (>2 cm) have a higher recurrence rate, ranging from 21% to 59%. Furthermore, Hürthle cell cancers do not always concentrate iodine. For these reasons, Hürthle cell carcinoma carries a worse prognosis compared with DTC. Adenomas have an excellent prognosis after resection and less than 2.5% demonstrate malignant behavior, but resection is recommended for larger adenomas because size is a major predictor of malignancy.

As follicular or papillary cancers progress or dedifferentiate, their prognosis becomes much worse. Anaplastic cancers are at the least differentiated end of the spectrum and represent one of the most aggressive cancers, with 5-year disease-free survival and cause-specific survival rates of 0%. ATC can arise from well-differentiated tumors, or it can also develop de novo (see Fig. 68.1C ). A group of tumors falls between well-differentiated thyroid cancers and anaplastic cancers. These cancers, called poorly differentiated thyroid cancers, are intermediate in terms of their histologic appearance and their biologic behavior. Although the literature remains inconsistent about what constitutes poorly differentiated cancer, the best definition comes from Burman and colleagues: “poorly differentiated thyroid carcinoma is a concept proposed to include carcinomas of follicular thyroid epithelium that retain sufficient differentiation to produce scattered small follicular structures and some thyroglobulin (Tg), but generally lack the usual morphologic characteristics of papillary and follicular carcinoma.” These tumors include insular, large cell, tall cell, columnar cell, solid, and diffuse sclerosing variants. In patients with these variants, the cancer tends to recur and metastasize. Furthermore, dedifferentiation of thyroid cancers leads to underexpression or disordered assembly of the sodium-iodide symporter, decreasing the usefulness of RAI for treating micrometastatic disease or detection of metastases. For these reasons, poorly differentiated thyroid cancers have a 51% disease-free survival rate and a 70% cause-specific survival rate at 5 years.

Primary lymphoma of the thyroid is not as common as DTC. Older women or patients with Hashimoto thyroiditis are at highest risk for developing thyroid lymphoma. These tumors typically manifest as a rapidly expanding mass causing pain and compressive symptoms. Flow cytometry of cytologic specimens can sometimes be used to make the diagnosis, but the condition might be mistaken for Hashimoto thyroiditis. Consequently, a core biopsy is sometimes necessary when this diagnosis is suspected. Most are B-cell lymphomas treated with chemotherapy and radiation. Surgery is occasionally needed for palliation. Prognosis depends on the histologic subtype.

Diagnosis

Just as with a newly discovered mass anywhere else in the body, the workup of a thyroid nodule begins with a thorough history and physical examination. A strong family history of thyroid cancer or cancer syndromes or a history of radiation exposure to the head and neck or total-body radiation for bone marrow transplantation should raise the suspicion of thyroid cancer. Rapid growth and/or hoarseness with compressive symptoms may indicate that the thyroid nodule is thyroid lymphoma or a poorly differentiated thyroid cancer. On examination, malignant nodules are harder and fixed, whereas a nodule that is rubbery or soft and moves easily with deglutition is reassuring but not diagnostic of a benign nodule. Nonpalpable nodules have the same risk of malignancy as palpable nodules of the same size. Cervical lymphadenopathy also increases the likelihood that a thyroid nodule is malignant.

Laboratory Studies

Because the management of patients with functional thyroid nodules differs from that of patients with nonfunctional nodules, obtaining a thyroid-stimulating hormone (TSH) measurement early in the workup of a thyroid nodule can efficiently identify patients with a nodule and hyperthyroidism. In this subset of patients with a suppressed TSH, an iodine-123 ( 123 I) scan can distinguish a solitary toxic nodule from a toxic multinodular goiter and Graves disease. A solitary hyperfunctioning nodule is rarely malignant, and fine-needle aspiration (FNA) biopsy or further cancer workup is rarely necessary. The one exception is that functioning nodules in children do carry a higher risk of malignancy. If thyroid radionuclide scanning is undertaken, “cold” nodules should undergo FNA biopsy because 10% to 20% of cold nodules are malignant. A higher serum TSH is an independent risk factor for predicting malignancy and is associated with more advance disease.

Other laboratory tests can be helpful once the diagnosis of a certain type of thyroid cancer has been made. For example, measuring serum Tg in patients with DTC can assist with the long-term follow-up of patients treated for DTC. Although Tg levels can be elevated in patients with DTC, the test is insensitive and nonspecific for diagnosing cancer, elevations in Tg can occur in benign thyroid disorders, and the American Thyroid Association (ATA) guidelines do not recommend routine preoperative Tg measurement for patients with DTC. After a total thyroidectomy, however, elevations in Tg can reliably indicate recurrent or metastatic disease. Different threshold Tg levels can indicate recurrence depending on the concomitant TSH level. It should be emphasized, however, that there is no role for Tg measurement in the initial evaluation of thyroid nodules.

Measurement of serum anti–thyroid peroxidase (TPO) antibodies may be helpful in patients with high TSH levels suggestive of autoimmune thyroiditis; however, routine measurement is not necessary.

Fine-Needle Aspiration Biopsy

FNA biopsy remains the gold standard for evaluating thyroid nodules. Most clinical practice guidelines recommend FNA biopsy for nodules greater than 1 cm in largest dimension with a high to intermediate sonographic pattern. Biopsy can be performed on nodules larger than 1.5 cm with few suspicious characteristics. For nodules larger than 1 cm with suspicious sonographic features of extrathyroidal extension or associated lymphadenopathy, FNA should be performed in selected patients such as those with family history of thyroid cancer. When the FNA result is clearly benign or malignant, then the decision for further treatment, including thyroidectomy, becomes evident. The false-negative rate for FNA biopsy is 1% to 3% ( Table 68.5 ). The false-negative rate increases to 10% to 15% when the nodule is large (>4 cm). Other clinical scenarios in which the clinician should not always trust a benign FNA result include patients with a family history of thyroid cancer, patients with a history of radiation exposure, and cystic nodules. Ultrasound guidance can improve the accuracy of FNA biopsy by confirming that the nodule is actually being sampled and by enabling targeting of the most suspicious portions of the nodule (e.g., the wall of a cyst). This is especially true for nondiagnostic cytologic findings (>25%–50% cystic component) or nonpalpable or posteriorly located nodules.

Table 68.5
The Bethesda System for Thyroid Cytopathology
Category Risk of Malignancy (%) Recommended Management
Nondiagnostic or unsatisfactory 1–4 Repeat FNA with ultrasound guidance
Benign 0–3 Clinical follow-up
Atypia of undetermined significance (AUS) or follicular lesion of undetermined significance (FLUS) 5–15 Repeat FNA a
Follicular neoplasm or suspicious for follicular neoplasm 15–30 Lobectomy
Suspicious for malignancy 60–75 Lobectomy with or without frozen section or total thyroidectomy
Malignant 97–99 Total thyroidectomy
FNA, Fine-needle aspiration.

a Lobectomy also can be considered depending on clinical or sonographic characteristics.

Nodules that do not meet FNA criteria, such as suspicious subcentimeter nodules in individuals older than 60 years, single nodules with well-defined margins, and nodules with a rim of normal thyroid parenchyma that is larger than 2 mm, can be observed.

FNA results are classified according to the Bethesda criteria, which indicate the risk of malignancy (see Table 68.5 ). One of the limitations of cytologic evaluation of thyroid nodules is that it cannot distinguish between adenoma and carcinoma in follicular lesions. Therefore lobectomy with permanent histology may be the best way to make a definitive diagnosis in follicular or indeterminate lesions. Furthermore, 20% to 30% of FNA results fall into the category of indeterminate cytologic findings, such as atypia of undetermined significance (AUS) or follicular lesion of undetermined significance (FLUS). Currently, many centers have turned to molecular analysis of FNA specimens to help distinguish follicular lesions. Cytologic specimens are analyzed for a panel of mutations, including BRAF, RAS, RET/PTC, and PAX8-PPARγ rearrangements. The seven-gene mutational panel, most useful when surgery is favored, has sensitivity that ranges from 44% to 100%. Although this is an exciting area of research, the clinical usefulness of the various gene panels has varied, and more prospective data are needed.

Testing for molecular markers from FNA aspirates could be conducted for FLUS, AUS, or follicular neoplasm. These are BRAF and RAS mutational status, high-density genomic data for molecular classification (an FNA-trained mRNA classifier), and FNA-trained miRNA classifier combined with molecular markers of malignancy. Patient counseling regarding the potential benefits and limitations and the long-term clinical implications of the molecular testing should be done according to the ATA guidelines. The guidelines also recommend that these tests should be performed in Clinical Laboratory Improvement Amendments/College of American Pathology (CLIA/CAP)–certified molecular laboratories or the international equivalent.

Imaging

Not only can ultrasonography improve the accuracy of FNA biopsy, but it is also an important tool in evaluation of thyroid nodules because it is used to measure the size and features of the nodule and it can also reveal additional nonpalpable nodules. Ultrasound examination alone can increase the clinician's suspicion for malignancy if the nodule has fine microcalcifications, irregular borders, chaotic vascular patterns or peripheral vascularity, cystic aspect, or hyperechogenicity. In addition, ultrasound scanning can be used to evaluate the lymph nodes in both the central and the lateral neck compartments, which may prompt additional FNA biopsy of suspicious lymph nodes or result in alterations in the surgical plan. Although ultrasound examination is highly operator dependent, it is noninvasive and does not involve any radiation or contrast agent risk to the patient. High-resolution ultrasonography can also demonstrate extracapsular invasion and subtle lymph node involvement. Consequently, ultrasonography is the preferred method to evaluate the thyroid and cervical lymph nodes. Although routine screening is not recommended for all patients, those with a strong family history or radiation exposure can undergo ultrasound screening for thyroid nodules. Use of positron emission tomography (PET) and/or computed tomography (CT) scan is helpful for identifying lung or bone tumors in patients at risk for metastases.

Highly aggressive cancers that may invade local structures, extend into the chest, or demonstrate poorly differentiated cytologic features require careful preoperative planning. In such cases, CT becomes a helpful preoperative imaging study in planning en bloc resection of other organs aside from the thyroid, understanding the extent of vascular involvement, determining if a thoracic incision is necessary, and planning for reconstruction.

Chest CT or PET-CT imaging studies performed for other medical indications often reveal thyroid nodules incidentally. PET scans demonstrate thyroid masses during the workup and staging of other cancers. This subset of incidentally discovered thyroid nodules deserves special attention because up to 50% of fluorodeoxyglucose (FDG)-avid thyroid nodules will contain thyroid cancer. Therefore, PET-positive thyroid nodules should undergo FNA biopsy.

Treatment

Treatment of DTC involves a surgeon, an endocrinologist, a nuclear medicine specialist, and, occasionally, a radiation oncologist. A multidisciplinary approach best serves patients with DTC.

Surgery

The extent of surgery for DTC remains controversial. This is especially true for small, encapsulated, well-differentiated tumors, and tumors smaller than 1 cm (microcarcinomas). These are discussed further later, but for most DTCs 4 cm or larger that were diagnosed preoperatively, most clinicians recommend a total thyroidectomy. Controversy still exists for lesions larger than 1 cm and smaller than 4 cm with low- or intermediate-risk features. Contrary to the previous guideline recommendations, the new guidelines recommend either total thyroidectomy or lobectomy for treatment. Thyroid lobectomy may be sufficient for this type of lesion without extrathyroidal extension and with no evidence of lymph node metastasis. This is based on evidence from properly selected patients that showed similar clinical outcomes with either surgical plan. Furthermore, lobectomy can obviate the need for exogenous thyroid hormone. Finally, because the current practice for follow-up depends more on ultrasound findings and Tg measurement than on whole-body RAI, total thyroidectomy is no longer needed to justify postoperative RAI. Analysis of 5432 patients with PTC from the SEER database found no difference in 10-year overall survival between total thyroidectomy (4612 patients) and thyroid lobectomy (820 patients). Results from two other single-center studies showed excellent survival in properly selected patients who underwent lobectomy. Without evidence of high-risk features, initial lobectomy of 1- to 4-cm thyroid carcinomas is more cost-effective after 3 years of follow-up. On the other hand, the rationale for total thyroidectomy is based on tumor biology and current treatment modalities. DTC, especially PTC, tends to be multicentric, with up to 80% of patients having multiple tumor foci and 60% having bilateral disease when a thorough pathologic examination of the contralateral lobe was performed. A study showed that 43% of patients who underwent thyroidectomy for 1- to 4-cm thyroid cancers had high-risk characteristics that would have necessitated complete thyroidectomy if lobectomy had been used as initial treatment. A total thyroidectomy as the initial procedure obviates the need for reoperative surgery to remove the contralateral lobe should a recurrence become detected. Second, experienced thyroid surgeons can safely perform a total thyroidectomy, with permanent complications such as recurrent laryngeal nerve injury and hypoparathyroidism occurring at a rate of less than 2%. RAI therapy for ablation of microscopic disease becomes most effective when the thyroid remnant is small or absent. Tg measurement and radioiodine whole-body scanning are highly sensitive modalities for detection of recurrent or metastatic disease, but these two methods are most effective when no thyroid tissue remains in the neck.

Most low-risk cancers carry an excellent prognosis regardless of the extent of thyroidectomy, and there are no randomized prospective trials comparing total thyroidectomy and thyroid lobectomy in this group of patients. In addition, radioiodine may have limited usefulness in low-risk patients. For these reasons, some researchers favor thyroid lobectomy in low-risk patients. For example, Shaha and colleagues have reported 20-year follow-up findings in 465 patients with low-risk DTC. Although the lobectomy group had more local recurrence compared with the total thyroidectomy group (4% versus 1%), there was no statistical significance. Similarly, other groups have also failed to demonstrate any significant effect on survival. In contrast, large retrospective series have demonstrated improvement in recurrence rates for total thyroidectomy compared with less extensive operations. In a frequently cited study, Mazzaferri and colleagues reported on 1355 patients with a mean follow-up of 15.7 years. Patients treated with total thyroidectomy experienced significant improvements in recurrence rate (26% versus 40%, P < .02) and mortality rate (6% versus 9%, P = .02) compared with less extensive resections. Owing to concerns regarding the accuracy of risk stratification and complications in these retrospective studies, current guidelines recommend lobectomy for small (<4 cm), unifocal, well-differentiated tumors with no lymph node metastases or extrathyroidal extension.

Another hotly debated topic related to the extent of initial surgery for DTC is the role of prophylactic central neck dissection. The most recent guidelines are consistent with the 2009 guidelines, which state that “prophylactic central neck dissection may be performed, especially in patients with advanced primary tumors (T3 or T4) or clinically involved lymph nodes” and “total thyroidectomy without prophylactic central neck dissection may be appropriate for small (T1 or T2), noninvasive, clinically node-negative patients.”

The central neck lymph nodes are also classified as level VI lymph nodes and include the paratracheal, perithyroidal, and precricoid lymph nodes. These nodes are found along and behind the recurrent laryngeal node, and frequently surround the lower parathyroid gland. Although the level VI lymph nodes contain macroscopic disease in 10% of cases, when they are removed prophylactically, 32% to 69% of patients will have microscopic metastases.

Proponents of prophylactic central neck dissection argue that the initial operation is the safest time to remove central neck lymph nodes to prevent local recurrences and the complications associated with reoperative surgery in the central neck. Wada and colleagues found the recurrence rate in patients treated with therapeutic lymph node dissection to be 21%, whereas patients who underwent prophylactic neck dissection experienced a recurrence rate of only 0.43%. Important to note, those patients without clinically overt nodal disease who did not undergo prophylactic central neck dissection also experienced a very low recurrence rate of 0.65%. Hence, the absolute differences in recurrence are minuscule. Several other studies also have supported the concept that microscopically positive lymph nodes rarely progress to recurrence, especially after postoperative RAI ablation. Clinically evident lymph node metastases place patients at higher risk for recurrence, and these patients clearly benefit from therapeutic lymph node dissection. Prophylactic central neck dissection reduces an already low recurrence rate and potentially eliminates or reduces the need for RAI but is also associated with risks such as hypoparathyroidism. The risk-to-benefit ratio may favor prophylactic central neck dissection in a subset of patients, but the putative risk factors that define such a subset remain unknown.

Evidence supports a selective approach of prophylactic level VI node dissections in patients with clinically or radiographically involved lymph nodes or intraoperative detection of metastatic lymph nodes (cN1). Thyroidectomy begins with proper patient positioning in the semirecumbent position with the neck extended ( Fig. 68.2 ). A transverse, curvilinear incision is made in a suitable skin crease at or beneath the level of the cricoid cartilage. Traditionally a Kocher collar incision was used, but this requires a very large dissection superiorly to reach the upper pole of the thyroid, placing the patient at risk for postoperative seroma. Intraoperative ultrasound guidance can help in assessing the upper extent of the gland and placing the incision appropriately. Superior and inferior subplatysmal flaps are raised to create a working space around the thyroid. The strap muscles are divided in the median raphe and are retracted laterally. It is rarely necessary to transect the strap muscles, but this can be accomplished if the tumor is large or adherent to the overlying muscles. The perithyroidal soft tissue is swept off the gland bluntly to identify the boundaries of the thyroid. Because the thyroid is most fixed at the upper pole, these vessels are divided first. Much of this can be accomplished by using energy devices such as the Harmonic scalpel or LigaSure, but in larger vessels clips and/or ties may be required. The upper pole vessels should be ligated close to the thyroid capsule to avoid injuring the external branch of the superior laryngeal nerve. In addition, the surgeon should remain vigilant for the upper parathyroid gland, which is frequently located near the upper pole vessels. Next, the thyroid gland is reflected medially. To accomplish this, the middle thyroid vein is ligated. In addition, dividing the thyroid isthmus can also facilitate medial rotation, assuming that the tumor is not located within the isthmus. Before any structures along the medial border of the gland are divided, the recurrent laryngeal nerve must be identified and its course dissected. The nerve is found medial to the upper parathyroid gland and lateral to the lower parathyroid. The parathyroid glands must also be identified and dissected free from the thyroid on an intact vascular pedicle. Once the recurrent laryngeal nerve has been identified, the branches of the inferior thyroid artery can be divided along the thyroid capsule. The inferior pole also is mobilized with a combination of blunt dissection and ligation of the inferior thyroid artery. The thyroid is then dissected off the anterior surface of the trachea with electrocautery or other energy devices to divide the small vessels contained within the ligament of Berry. Performing the identical procedure on the contralateral lobe completes a total thyroidectomy.

Figure 68.2, Thyroidectomy. (A) The patient is placed with the neck in extension. The thyroid is approached through a Kocher collar incision, which is commonly made approximately 2 cm superior to the sterna notch. (B) The strap muscles are divided in the midline to expose the thyroid gland. (C) The strap muscles are retracted laterally and the thyroid is retracted medially, exposing the structures of the midneck. The recurrent laryngeal nerve can be seen lying within the tracheoesophageal groove. (D) The superior pole vessels are individually clamped and ligated as they enter the thyroid gland. Inferior thyroid vessels, in addition to the thyroidea ima vessels, are individually suture-ligated. (E) The dissection is completed by dissection of the thyroid gland off the trachea. The isthmus is then transected and can be oversewn with a suture for hemostasis.

Before passing the specimen off the field, the surgeon should examine it to make sure that there is no parathyroid tissue adherent to the gland. Any inadvertently removed parathyroid tissue can be finely minced and reimplanted into either the sternocleidomastoid or the strap muscles. Frozen section of a biopsy specimen of this tissue can be used to distinguish among fat, parathyroid, or lymph node; this will also help in avoiding autotransplanting cancer-bearing lymph nodes back into the patient. Two or three pockets are created within the muscle, and the minced parathyroid tissue is divided among these pockets. Each pocket should be marked with permanent suture so that it can easily be found in a reoperative setting.

Preoperative FNA or intraoperative frozen section can be used to confirm that enlarged lymph nodes seen on ultrasound harbor metastatic disease. Cytologic or pathologic confirmation of lymph node metastases should prompt the surgeon to perform a compartment-oriented lymph node dissection. Lymph node sampling or “berry-picking” should be avoided, because this leaves behind lymph nodes that likely contain microscopic disease, which then become more difficult to excise in a reoperative setting.

Although “skip” metastases directly to the lateral compartment can occur in PTC, the central neck nodes (level VI) are usually the first nodes to receive drainage from the thyroid ( Fig. 68.3 ). The boundaries of the central neck are the carotid sheathes laterally, the hyoid bone superiorly, and the innominate artery inferiorly. Lymphadenectomy in this area requires skeletonizing the recurrent laryngeal nerve along its entire cervical course, and removing all the fibrofatty tissue along the trachea. Frequently, the lower parathyroid is invested in this tissue and becomes devascularized with this dissection.

Figure 68.3, The central neck nodes (level VI) are usually the first nodes to receive drainage from the thyroid.

A lateral neck dissection usually involves dissection of levels II, III, and IV (see Fig. 68.3 ). This dissection puts the spinal accessory, phrenic, vagus, cervical sensory, sympathetic trunk, hypoglossal, greater auricular, and marginal mandibular branch of the facial nerves at risk. The extent of node dissection should be guided by preoperative and intraoperative ultrasound findings. Usually the great vessels can be preserved, but more aggressive tumors can invade the internal jugular vein, and it should be sacrificed in this scenario. In addition, to nerve injury, chyle leak is another complication of lateral neck dissection.

Radioactive Iodine

Remnant ablation with RAI is the standard adjuvant treatment for selected patients with DTC. Because the primary goal of RAI is remnant ablation, it can be administered only after a total or near-total thyroidectomy; otherwise the radioactive isotope will be absorbed by the remnant thyroid and will not destroy any micrometastatic disease as intended. In addition, RAI can be used as an adjuvant therapy after total thyroidectomy to improve disease-free survival or as a therapeutic modality to improve survival in patients with persistent disease. RAI is administered 1 to 3 months postoperatively as iodine-131 ( 131 I) as sodium iodide in an oral form whose half-life is 7 to 8 days ( Figs. 68.4 and 68.5 ). Consensus guidelines recommend a dose of 30 to 100 mCi for patients with low-risk tumors and higher doses (100–200 mCi) for patients with residual disease, suspected microscopic disease, or more aggressive histologic subtypes (i.e., tall cell, columnar cell, or insular variants). To stimulate intracellular uptake of the isotope, the TSH concentration should be at least as high as 30 mU/L. There are two methods for achieving such an elevation in TSH. The traditional method requires the patient to withdraw from thyroid hormone replacement over 4 to 6 weeks. A newer method is to administer recombinant human thyroid-stimulating hormone (rhTSH). rhTSH is administered in the form of intramuscular injections on 2 consecutive days followed by RAI on the third day. The advantage of this method is that the patient does not experience an extended period of hypothyroidism as with hormone withdrawal. However, long-term data on the effectiveness of rhTSH compared with traditional withdrawal are lacking, although it appears effective for low-risk and intermediate-risk patients; there is no available evidence to support rhTSH use in patients with high risk of disease-related mortality and morbidity. The US Food and Drug Administration (FDA) approved rhTSH for thyroid remnant ablation in patients who do not have evidence of metastatic disease. In addition to increasing the TSH level, clinicians should also prepare patients by instructing them to follow a low-iodine diet for 1 to 2 weeks before RAI treatment. This diet requires patients to avoid foods that contain iodized salt, dairy products, eggs, seafood, soybeans or soy-containing products, and foods colored with red dye No. 3.

Figure 68.4, (A) Whole-body scan acquired 24 hours after administration of 2 mCi of iodine-123 ( 123 I). (B) Spot view of the neck and chest. There are several areas of uptake indicative of residual thyroid and functioning metastases in cervical lymph nodes. (C) Posttherapy scan made 7 days after administration of 150 mCi of 123 I. There is intense uptake in the region, but the resolution is not as good as with 123 I. There is faint uptake in the liver on the posttreatment scan as a result of metabolism of radioiodinated thyroid hormones at that site.

Figure 68.5, Positron emission tomography (PET) scan of a 75-year-old woman with anaplastic cancer of the thyroid. Images were acquired 1 hour after intravenous injection of 15 mCi fluorine-18 fluorodeoxyglucose (FDG). There is intense uptake of FDG in the undifferentiated cancer.

Although some studies have shown no benefit to RAI therapy, other studies have demonstrated a reduction in locoregional recurrences and distant metastases. As with the controversy over the extent of thyroidectomy, the benefit of RAI for low-risk patients remains unclear. The most recent ATA (2016) guidelines recommend remnant ablation for all but the lowest-risk patients (unifocal, well-differentiated tumor, <1 cm in size, confined to the thyroid gland without lymph node metastases or multifocal papillary carcinoma with no other adverse findings). These recommendations are supported by multiple systematic reviews including one that was published in 2015. In these low-risk patients, most of the available evidence suggests that RAI does not improve disease-specific or disease-free survival. The National Comprehensive Cancer Network (NCCN) guidelines require a more thorough evaluation for the extent of remaining disease after thyroidectomy, with a radioiodine scan 1 to 12 weeks postoperatively. RAI ablation is not recommended if the stimulated Tg is less than 1 ng/mL and the radioiodine scan result is negative.

Some studies have shown an increase in the risk of development of secondary malignancies after RAI therapy. This has been examined with use of the National Cancer Institute's SEER database. Brown and colleagues found that patients treated for DTC had significantly higher rates of nonthyroid second primary malignancies than expected in the general population. Although the excess risk was relatively small, it was greater in the subset of patients who were treated with RAI. Iyer and colleagues specifically examined low-risk patients (T1N0) treated with RAI and found that their excess absolute risk was 4.6 excess cases per 10,000 person-years at risk. As discussed earlier, RAI clearly benefits patients with larger tumors and metastatic disease, but the increased risk of secondary malignancies in low-risk patients in whom the long-term benefit of RAI is questionable means that careful patient selection for RAI treatment is necessary.

Hematologic malignancies are the most common secondary malignancies after RAI, but there is also an association with kidney, breast, bladder, skin, and salivary gland cancers. The more commonly noted side effects after radioiodine treatment include dry mouth, mouth pain, salivary gland swelling (sialadenitis), altered smell and taste, conjunctivitis, and fatigue. Women should not be pregnant at the time of treatment, nor should they become pregnant for at least 6 months after treatment. Similarly, men should avoid conception for at least 6 months after treatment. Although a study has shown that BRAF V600E mutations significantly reduce sodium-iodine symporter expression and RAI uptake, there is no clear role of molecular testing in guiding postoperative RAI.

Thyroxine Suppression

Because all cells of follicular origin depend on TSH for growth, TSH suppression through the administration of supraphysiologic doses of levothyroxine (T 4 ) remains an important strategy for maintaining disease-free survival and overall survival. For high-risk patients with incomplete resection, tumor invasion into adjacent structures, or distant metastases, the physician should initially titrate T 4 dosage to a TSH level below 0.1 mU/L. Lower-risk patients should be treated to achieve a TSH level at or slightly below the lower limit of normal (0.1–0.5 mU/L). Once patients remain disease-free for at least 2 years, their TSH suppression can be liberalized to within the reference range. Patients with persistent disease should be kept at a TSH level below 0.1 mU/L indefinitely. TSH suppression carries risks of arrhythmias, anxiety, and osteoporosis. The risks and benefits should be carefully considered, particularly in older patients. Because of the risk of bone loss, the NCCN guidelines recommend daily calcium and vitamin D supplementation for patients on TSH suppression.

External Beam Radiation

Although 131 I is the preferred adjuvant therapy for thyroid carcinoma, external beam radiation sometimes plays a role in treating this disease. Persistent, recurrent, anaplastic, or poorly differentiated tumors may fail to take up 131 I. Treatment of anaplastic thyroid tumors almost always includes external beam radiation because these tumors often cannot be completely resected and do not concentrate iodine. Although no improvement in overall survival has ever been documented, external beam radiation is often given after resection of poorly differentiated tumors to reduce the risk of local relapse. The group at Memorial Sloan Kettering Cancer Center has found that up to 85% of poorly differentiated tumors display some iodine avidity, and therefore treatment with RAI may remain worthwhile. Patients with incompletely resected tumors, unresectable disease, and locoregional recurrence in a previously operated field may benefit from external beam radiation.

Chemotherapy

Because RAI often can be effective treatment for well-differentiated tumors that have metastasized, cytotoxic chemotherapy has not been extensively evaluated for metastatic thyroid cancers. For large burdens of disease, anaplastic cancers, or poorly differentiated tumors that are not iodine avid, chemotherapy becomes an important treatment component after surgery or if the tumor is not resectable. In these situations, chemotherapy confers minimal effects because these tumors carry a very poor prognosis. Historically, doxorubicin was the most effective single agent. Combination therapy with doxorubicin and cisplatin resulted in modest objective response rates. Newer, targeted therapies have shown some promise. Small-molecule tyrosine kinase inhibitors (such as sorafenib or sunitinib) and antibodies (anti–vascular endothelial growth factor [VEGF]) should be considered in the context of ongoing clinical trials. Sorafenib and lenvatinib have been approved for use in the United States for patients with advanced RAI-refractory DTC. Although there was no improvement in overall survival with sorafenib and lenvatinib, both were associated with prolongation of progression-free survival (PFS) by 5 months.

Recurrence

For most DTCs, the long-term survival rate exceeds 95%. Even though disease-specific mortality rates remain quite low (<1%), recurrence rates exceed mortality rates. For low-risk tumors, the locoregional recurrence rates range from 2% to 10% and distant recurrence rates are 1% to 2%. Higher-risk tumors (larger size, extrathyroidal extension, cervical lymph node metastases) carry recurrence rates of 21% to 68%. For these reasons, a tailored approach to follow-up and treatment of recurrent disease best serves patients with DTC.

Surveillance

The original tumor characteristics and operative findings dictate the follow-up schedule for DTC. Surveillance consists of measuring serum Tg, TSH, and anti-Tg antibodies in addition to imaging. Cervical ultrasound is a highly sensitive test for detection of metastatic lymphadenopathy. Elevations in Tg in the absence of cervical disease seen on ultrasound suggest distant metastases. Whole-body radioiodine scanning is sensitive for detecting iodine-avid bone or pulmonary metastases. Poorly differentiated, aggressive tumors will not concentrate iodine but can be detected with CT scan or FDG-PET (see Fig. 68.5 ).

The frequency of surveillance depends on the original tumor characteristics and the AJCC stage. For lower-risk tumors, physical examination, cervical ultrasound, and measurement of TSH, Tg, and anti-Tg antibodies should be performed every 12 months. These studies can be scheduled 3 to 6 months after the initial RAI treatment in patients with high-risk tumors.

Treatment of Recurrent Disease

Recurrence in the neck or cervical lymph nodes is best treated surgically. The decision-making process of surgical resection should be based on clinically apparent, macroscopic nodal disease confirmed with neck ultrasound or CT scanning, rather than depending on Tg elevation alone. Furthermore, small nodal recurrence (<8 mm in the central neck or <10 mm in the lateral neck) can be managed with active surveillance.

Other treatment options such as percutaneous ethanol injection (PEI) have shown some success in retrospective studies limited by small sample sizes. External beam radiation should be considered for recurrent disease that is unresectable or not iodine avid ( Fig. 68.6 ). Percutaneous ultrasound-guided laser ablation has shown promising results for metastatic nodal disease. RAI cannot ablate bulky nodal disease. After surgical resection, radioiodine can effectively ablate micrometastatic disease throughout the body.

Figure 68.6, External beam radiation for thyroid carcinoma can be quite complex. On the left, the target for this bulky thyroid carcinoma is marked with a dashed line. The high-dose volume (95% dose line) encompasses this target while avoiding the spinal cord. The radiosensitive spinal cord is in the 60% isodose line, which permits delivery of doses up to 70 Gy with this plan. On the right is a superimposition of the six cross-firing fields that are used to create this dose distribution. The fields either avoid the spinal cord or include a lead block to shadow the spinal cord, protecting it from the high-dose radiation. The treatment planning and dosimetry of thyroid carcinoma treatment is one of the most complex challenges in radiation oncology.

Medical treatment for recurrent or metastatic disease consists of maintaining TSH suppression. Depending on the location of recurrence, health of the patient, tumor risk stratification, and patient preference, patients with metastatic disease can be referred for experimental protocols using targeted therapies, can undergo traditional cytotoxic chemotherapy, or can be treated with watchful waiting and supportive care. A multidisciplinary team experienced with metastatic thyroid cancers best handles these decisions.

Medullary Thyroid Cancer

The prevalence of MTC has decreased from previously reported rates of 5% to 10% of all thyroid cancers to 1% to 2%. This is because of the increased incidence of PTC. Unlike DTC, MTC arises from the parafollicular C cells instead of the follicular epithelium (see Fig. 68.1D ). Hence, MTC is a neuroendocrine tumor (NET), and it shares some properties common among neuroendocrine cancers, including secretion of peptide hormones such as calcitonin, serotonin, or vasoactive intestinal peptide (VIP). Most cases of MTC are sporadic, but 25% are a result of germline genetic mutations. Hereditary cases occur either in isolation (familial medullary thyroid carcinoma [FMTC]) or as part of MEN syndrome type 2 (MEN2A or MEN2B).

Diagnosis

Although any thyroid nodule could potentially harbor MTC, historical features that may alert the physician to the potential for MTC include a family history of MTC, PCC, hyperparathyroidism, or other manifestations of MEN2 syndromes. The median age of diagnosis is approximately 50 years. In a SEER study that included 1252 patients between 1973 and 2002, the authors found that 87% of the cohort were white and 60% were female. As in evaluating all thyroid nodules, neck ultrasound examination and FNA play a major role in diagnosing MTC. In one retrospective study, ultrasound was found to be falsely negative for ipsilateral and central node involvement in 17% and 14%, respectively, of patients with MTC with no ultrasound pathognomonic features for MTC. Hereditary cases are often detected through genetic screening to identify germline mutations in the RET gene. Almost all sporadic cases manifest with a palpable neck mass, which could be either the thyroid mass or a metastatic lymph node. In contrast to sporadic disease, hereditary disease usually manifests as multicentric and bilateral and involves the upper part of the thyroid. However, bilateral disease in sporadic MTC with negative RET mutation can occur in up to 9% of patients. Lymph node metastases occur in 35% to 50% of patients at initial diagnosis. Therefore, ultrasound evaluation of the central and lateral neck compartments for suspicious lymph nodes becomes a crucial component to the initial diagnosis.

Because the parafollicular C cells are concentrated in the upper, posterior portion of each thyroid lobe, many MTCs arise in a posterior location, causing symptoms such as hoarseness or dysphagia as a result of compression of local structures. If there is any concern for vocal cord function, then direct laryngoscopy should be performed preoperatively. Markedly elevated calcitonin levels can cause symptoms such as flushing, diarrhea, and weight loss. Distant metastasis to the liver is the most frequent site. CT scan is a very sensitive imaging modality for detection of local nodal and distant lung metastases. Liver metastases are best detected with MRI, and routine use of fluorine-18 fluorodeoxyglucose ( 18 F-FDG) PET-CT is not recommended in MTC evaluation because it is less sensitive.

FNA sensitivity in detecting MTC ranges from 50% to 80% and can be improved with the addition of immunohistochemical staining of calcitonin. A meta-analysis of 15 publications showed an accuracy of FNA of less than 50%. FNA characteristics of MTC include the presence of stromal amyloid without thyroid follicles. Because spindle-shaped cells may be seen, MTC can be mistaken for parathyroid carcinoma or ATC unless the specimen is stained for calcitonin, chromogranin A (CgA), or carcinoembryonic antigen (CEA)—substances produced by MTC that confirm the diagnosis. A more sensitive technique than immunohistochemistry on cytology specimens is to measure the calcitonin level in the washout fluid from an FNA. In addition, the presence of calcitonin messenger RNA (mRNA) has been performed when the cytologic or histologic diagnosis remains unclear.

Several serum markers can confirm the diagnosis of MTC and are useful in following patients for recurrence and metastases. Calcitonin is commonly elevated in patients with MTC. Although a small percentage of normal patients will have some elevation in calcitonin, patients with a diagnosis of MTC typically exhibit levels above 100 pg/mL. In borderline cases, the diagnosis can be clarified by stimulating the calcitonin with either intravenous calcium gluconate or pentagastrin. Of note, calcitonin secreted by nonthyroid tissue such as lung cancer or prostate cancer does not elevate with calcium gluconate or pentagastrin stimulation. Currently available immunochemiluminometric assays (ICMAs) are more sensitive and specific for monometric calcitonin and largely eliminate cross-reactivity with procalcitonin.

Before the advent of genetic testing, these stimulated measurements were used to screen patients at high risk for MTC. The degree of calcitonin elevation correlates with tumor burden, with nodal metastases found with basal calcitonin levels of 10 to 40 pg/mL and distant metastases found with calcitonin levels greater than 150 pg/mL. Patients with calcitonin levels greater than 3000 pg/mL are likely to have widely metastatic disease and are unlikely to experience a cure.

Preoperative measurement of serum CEA can also help risk-stratify patients. Overall, CEA elevations occur in more than 50% of patients with MTC, but a preoperative serum CEA level greater than 30 ng/mL highly predicts the inability to cure the patient with surgery. CEA levels above 100 ng/mL may signify extensive lymph node and distant metastases. Following CEA levels postoperatively can also be used to monitor disease progression. Simultaneous increases in CEA level and calcitonin level indicate disease progression, whereas elevation of CEA level in the presence of a stable calcitonin level is associated with a worse prognosis because it may indicate tumor dedifferentiation and distant metastases. However, normal CEA or calcitonin level is rare and could represent an advanced dedifferentiated MTC or misdiagnosis. Other markers, such as CgA and serotonin, may be elevated in patients with MTC, as with many other NETs, but calcitonin and CEA are the most useful for following MTC patients in the long term.

Genetic testing plays an important part in the initial management because it can be used to identify familial disease and to risk-stratify patients. Germline mutations in the RET gene characterize familial disease. A small percentage of apparently “sporadic” disease will also carry germline RET mutations, but truly sporadic cases frequently harbor somatic RET mutations. For sporadic MTCs lacking RET mutations, 18% to 80% have been found to have HRAS, KRAS, or NRAS mutations. In sporadic MTC, presence of RET, especially RET codon M918T, mutation usually predicts a poor prognosis. Commercial testing is performed through polymerase chain reaction (PCR) amplification of the patient's germline DNA obtained from the patient's white blood cells. A spectrum of tumor aggressiveness exists among the various RET mutations, and the timing of prophylactic thyroidectomy is based on the specific mutation. Once a patient tests positive for a germline RET mutation, the patient should be carefully counseled regarding the risk to other family members and the patient's children. At-risk family members should be identified and also tested so that prophylactic thyroidectomy can be offered at the appropriate time. Although some overlap exists for genetic mutations associated with MEN2A and familial MTC, distinct mutations are usually associated with MEN2B.

Treatment

Complete surgical excision is the treatment of choice for MTC. The minimum extent of surgery for patients with clinically apparent disease is a total thyroidectomy with bilateral central neck dissection. Eighty-one percent of patients with palpable disease have central neck lymph node metastases, and the addition of central neck dissection improves cure rates over total thyroidectomy alone in patients with clinically evident disease at presentation. The involvement of ipsilateral and/or contralateral nodes has been found to be positively correlated with presence and number of central nodes metastasis. Furthermore, calcitonin level is helpful in preoperative risk assessment of nodal disease. No risk of lymph node metastasis was found when serum calcitonin level was less 20 pg/mL in a study of 300 patients with MTC. In this study, metastasis to the ipsilateral central neck, ipsilateral lateral neck, contralateral central neck, contralateral lateral neck, and upper mediastinum were associated with calcitonin levels greater than 20, 50, 200, and 500 pg/mL, respectively. The initial approach to lateral neck lymph nodes continues to evolve. Historically, the initial surgical treatment included an ipsilateral lateral compartment neck dissection because up to 80% of patients will have ipsilateral nodal metastases. However, current guidelines recommend performing an ipsilateral lateral neck dissection if ultrasound or physical examination detects lymphadenopathy in the lateral neck, if central compartment lymph nodes are involved, or if the primary tumor is greater than 1 cm. Intraoperative assessment of nodal disease by surgeons has sensitivity of 64% and specificity of 75%. Contralateral lateral neck dissection is added when patients have bilateral tumors or there is extensive lymph node disease on the ipsilateral side. Because some patients require extensive neck dissection, these procedures are often staged. Unlike DTC, in which micrometastatic disease can be effectively treated with RAI ablation, the only effective treatment for MTC is complete surgical resection. Therefore all evident disease must be resected for the best chance of long-term cure. According to the revised ATA guidelines, complete thyroidectomy is not indicated unless the patient has a RET germline mutation, elevation of serum calcitonin level, or evidence of residual MTC on images.

Prophylactic thyroidectomy is recommended for at-risk family members in hereditary MTC. Current recommendations for the timing of prophylactic thyroidectomy balance the need to remove the at-risk organ before it develops clinically apparent disease with the risks of surgery. In hereditary MTC, an age-related progression exists from C-cell hyperplasia to carcinoma, and, ultimately, nodal metastases. The optimal timing of prophylactic thyroidectomy depends on the risk level of the RET mutation. In general, current guidelines recommend operating on children with MEN2A and familial MTC by age 5 years, whereas those with MEN2B should be operated on before 6 months of age. Prophylactic surgery should consist of at least a total thyroidectomy. The role of prophylactic lymph node dissection in familial disease remains controversial. Lymph node metastases are present in 6% of screened patients, and therefore some argue that prophylactic central lymph node dissection should be performed. Opponents to this approach state that with normal preoperative ultrasound findings, normal calcitonin level (basal and/or stimulated), and a normal CEA level, the risk of occult nodal disease is very low and does not outweigh the risks of a central neck dissection such as permanent hypoparathyroidism. Because any complications resulting from prophylactic surgery become lifelong problems for the patient, experienced surgeons should perform prophylactic surgery. Before proceeding with surgery, the surgeon should screen patients with hereditary disease for associated conditions such as PCC (MEN2A and MEN2B) and hyperparathyroidism (MEN2A).

All patients will require thyroid hormone replacement once the thyroid is removed, but TSH suppression is not required. After surgery, the next phase of treatment is surveillance. This begins 2 to 3 months postoperatively with a new baseline calcitonin and CEA measurement. If the calcitonin is undetectable, these patients can be followed with yearly calcitonin measurements. Imaging is undertaken when the calcitonin level rises.

A spectrum of disease severity exists for both hereditary and sporadic MTC, and therefore the natural history of MTC varies widely. Distant metastases in the lung, liver, or bone can arise and lead to death quite quickly. On the other hand, many patients live with a large tumor burden and very high calcitonin levels with few symptoms. Others develop intractable diarrhea. In this case, cytoreductive surgery or somatostatin analogues such as octreotide can palliate severe symptoms. Overall 10-year survival rates have been reported as 75% and 95% for tumor confined to the thyroid and regional stage disease, respectively, in a SEER registry study. In this study, age and stage were found to be stronger predictors of survival in univariate analysis. Furthermore, calcitonin doubling times of less than 6 months indicate worse prognosis, with 5-year and 10-year survival rates of 25% and 8%, respectively. Conventional chemotherapy regimens with doxorubicin, dacarbazine, capecitabine, and 5-fluorouracil (5-FU) have demonstrated limited efficacy in patients with MTC. According to the revised ATA guidelines, systematic chemotherapy should not be used as a first-line treatment. Newer, targeted therapies block the RET receptor tyrosine kinase or its multiple downstream pathways, such as the extracellular signal-related kinase (ERK), phosphatidylinositol 3-kinase (PI3-K)/Akt, p38 mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinase pathways. Some of these tyrosine kinase inhibitors inhibit multiple signaling pathways simultaneously. These targeted therapies have been evaluated in multicenter trials.

The FDA has approved one of these targeted therapies, vandetanib, for treatment of metastatic MTC. Vandetanib is a small-molecule inhibitor of the VEGF receptor, epidermal growth factor (EGF) receptor, and the RET tyrosine kinase. In a randomized controlled clinical trial, patients treated with vandetanib experienced a median PFS of 22.6 months compared with 16.4 months in patients treated with placebo. Another FDA-approved tyrosine kinase inhibitor is cabozantinib, which targets VEGFR1 and VEGFR2, c-MET, and RET. In a phase I/II trail of 35 patients with MTC, 17 patients had a partial response. Patients on tyrosine kinase inhibitors should be monitored for hypothyroidism. Important to note, none of these agents have been shown to prolong overall survival. More research is currently being conducted on other potential therapies.

Adrenocortical Cancer

Incidence

Adrenocortical cancer (ACC) is a rare endocrine malignancy with an incidence of 1 to 2 per million people; it causes less than 0.2% of all cancer deaths. After ATC, ACC is the second most aggressive endocrine cancer. Although ACC can develop at any age, it occurs in a bimodal age distribution: in patients younger than 5 years, and those in the fourth and fifth decades of life. In general, ACC in adults is more aggressive than in children, with rapid disease progression. Women are affected more than men, at a ratio of 1.5 : 1, and are more likely to have a functional tumor. The incidence of ACC also varies among countries. The highest incidence is in southern Brazil, with a 10 to 15 times higher incidence in children and with the majority of sporadic ACCs possessing a germline TP53 (R227H) mutation.

Pathogenesis

Most ACCs arise sporadically, and the pathogenesis is not completely understood. It is not known if ACC develops from hyperplastic or adenomatous adrenal nodules. However, several genetic alterations have been identified; the most common mutation involves overexpression of the insulin-like growth factor (IGF) gene. Next-generation sequencing in 1051 children with cancers revealed that 27 (69%) out of 39 patients with ACCs possessed a TP53 mutation. In this study, family history was not a predictor of an underlying predisposition syndrome in most patients. β-Catenin CTNNB1 was associated with poor outcome and decrease in overall and disease-free survival in patients with ACCs. Another potential tumor suppressor gene, ZNRF3, was identified in 21% of patients with ACCs with use of exome sequencing and single-nucleotide polymorphism (SNP) array analysis. Furthermore, tumors containing hypermethylated DNA have also been found to be associated with a poor prognosis. Other growth factor and growth factor receptor mutations have also been identified in sporadic ACC. Familial tumor syndromes that involve ACC include MEN1 (11q13), Li-Fraumeni syndrome (17p13), Beckwith-Wiedemann syndrome (11p15.5 and 15q11–13), and Carney complex (17q23–24 or 2p16).

Clinical Presentation

Most ACC patients are asymptomatic until the tumor reaches a size that causes compression of nearby structures, local invasion, or distant metastasis. Although symptoms are vague, they can include fever, early satiety, weight loss, pain, anemia, nausea, and fatigue. The presence of these symptoms in the setting of ACC is ominous. The mean duration from the onset of symptoms to the diagnosis of ACC varies from 6 to 16 months and appears to be independent of whether the tumor is functional.

In 40% to 60% of the patients, ACC results in increased hormone production (functional) that may cause symptoms and assist in correctly diagnosing these aggressive tumors. Because most ACCs are inefficient in mature steroidogenesis, they can secrete a variety of steroid precursors that result in clinical or subclinical syndromes. As many as 75% of the ACCs are associated with subclinical Cushing syndrome, which can be diagnosed with biochemical testing. Clinical Cushing syndrome with the classic symptoms may be present in up to 50% of the patients with functional ACC. Virilization caused by excessive androgen steroids or androgenic precursors is identified in approximately 20% of the patients, and the combination of clinical Cushing syndrome and virilization is present in 10% of the functional ACCs. Cushing syndrome with virilization almost always indicates ACC. Other clinical manifestations include feminization (5%–8%), hypoglycemia (<5%), and hypokalemic alkalosis (<5%). Men may have gynecomastia or low libido owing to estrogen overproduction by ACC. Eighty percent of children have virilization; 6% have isolated glucocorticoid excess.

Metastases are present in 20% to 50% of the patients with ACC at the time of diagnosis, with lungs (40%–50%), liver (40%), and lymph nodes (20%–30%) being the most common sites. Less common sites include the bones, spleen, pancreas, and diaphragm.

Diagnosis

The correct diagnosis of ACC is important for appropriate management; however, preoperative diagnosis of ACC is complex. At present, the diagnosis relies on clinical assessment, urinary and plasma biochemical tests, and imaging studies. Clinical suspicion should be raised in any patient with adrenal Cushing syndrome with an adrenal mass, increased urinary 17-ketosteroids, and age younger than 20 years, or in patients with an adrenal mass, increased urinary 17-ketosteroids, virilization or feminization, weight loss, anemia, or fever.

Hormonal workup includes plasma adrenocorticotropic hormone, cortisol, and urinary 17-ketosteroids, with or without a dexamethasone suppression test. Adrenal incidentalomas should also be evaluated with plasma or urinary metanephrines to rule out PCC, and with renin and aldosterone levels in patients with hypertension to rule out aldosteronoma.

The role of imaging in the evaluation of adrenal mass is valuable for assessing the tumor size and other specific imaging characteristics. It has been demonstrated that 92% of the ACCs are larger than 6 cm and that the risk of malignancy increases with adrenal tumor size as follows: 2% for tumors smaller than 4 cm, 6% for tumors that are 4 to 6 cm, and 25% for tumors larger than 6 cm. Furthermore, adrenal masses that increase in size within a 6-month period are also suggestive of malignancy. CT is the most useful imaging study to evaluate adrenal tumor size, adjacent organ involvement, and resectability. Irregular borders, heterogeneity, calcifications, radiodensity greater than 25 Hounsfield units (HU), delayed washout of less than 50%, and extension to the inferior vena cava all suggest malignancy. In the largest reported series evaluating CT imaging for ACC, the authors found that improved diagnostic accuracy could be obtained by selecting a radiodensity cutoff for adrenal lesions of 13.9 HU at CT imaging. Sensitivity was 100% and specificity was 68%. In addition, when performed correctly, intravenously administered contrast material washout of less than 50% carries a sensitivity and specificity of 100%. With gadolinium-enhanced magnetic resonance imaging (MRI), malignant lesions show rapid and marked enhancement and a slower washout pattern on T2-weighted images. Other suspicious features at MRI include peripheral enhancement, central necrosis, and internal hemorrhage. The overall MRI sensitivity and specificity are 81% to 89% and 92% to 99%, respectively.

Two other imaging modalities show encouraging results. The sensitivity of FDG-PET was reported to be 97% after a systematic review that included 21 studies for identifying ACC, with no difference reported between FDG-PET and FDG-PET/CT. C-metomidate-PET (MTO-PET) is another emerging adrenal imaging technique with similar accuracy. Another imaging technique is ( 123 I)iodometomidate (IMTO) scanning, which has the ability to reveal adrenal lesions with a sensitivity and a specificity of 89% to 85% and 38% to 100%, respectively.

FNA is a tool that should be discouraged for the diagnosis of ACC because it has a high false-negative rate and it cannot accurately distinguish a benign from a malignant adrenal mass. The definitive diagnosis of ACC is through pathologic evaluation. Grossly, these are large tumors that may invade the adjacent organs and large blood vessels, such as the inferior vena cava. The tumors are yellow to tan, with areas of necrosis and hemorrhage. In the absence of metastatic disease or local, capsular, or vascular invasion, the diagnosis of malignancy may be difficult. The Weiss criteria incorporate nine histologic features that help distinguish between malignant and benign tumors. These features include high nuclear grade (III or IV), mitotic rate greater than 5 per 50 high-power fields (HPFs), atypical mitoses, diffuse architecture, microscopic necrosis, 25% or fewer clear cells, capsular invasion, sinusoidal invasion, and venous invasion. A tumor with a score of 2 or lower is classified as benign, whereas a tumor with a score 3 or greater is considered malignant. Evaluation of molecular markers such as microRNAs, IGF2 overexpression, increased Ki-67, and specific genetic variations may prove to be a more accurate diagnostic tool. Ki-67 tumor expression greater than 10% positively correlates with a worse prognosis.

Treatment

The management of ACC should involve a multidisciplinary approach and is tailored to each patient's disease status.

Primary Disease

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here