Download as pdf or txt
Download as pdf or txt
You are on page 1of 45

D.

ENDOCRINE
68  Cancer of the Endocrine System
Ammar Asban, Anish J. Patel, Sushanth Reddy, Thomas Wang,
Courtney J. Balentine, and Herbert Chen

S UMMARY OF K EY P OI N T S
Thyroid Cancer • Cancer with well-differentiated order to assist in operative
• The incidence of thyroid cancer histologic features has an excellent planning.
is increasing, and there are 5-year survival rate (>95%). • Treatment begins with surgery. Most
approximately 33,500 new cases per • Older age and extent of invasion are thyroid cancers are treated with total
year in the United States. related to prognosis. thyroidectomy. Compartment-
• The incidence of differentiated • Lymph node involvement is oriented neck dissection is added
thyroid cancer is 14.3 per 100,000 associated with higher recurrence when there is metastatic disease in
people per year, with a female-to- but has questionable impact on the cervical lymph nodes.
male ratio of more than 3 : 1. survival. • Adjuvant therapy for differentiated
• Differentiated thyroid cancer (DTC) • Multiple staging systems exist for tumors is radioactive iodine
includes papillary thyroid cancer DTC. (iodine-131 [131I]).
(PTC), which accounts for 80% of all • For Hürthle cell adenoma, larger size • Patients must be prepared for
thyroid cancers; follicular thyroid (>6 cm) is predictive of malignancy. radioactive iodine ablation with low
cancer (FTC), which accounts for • For poorly differentiated tumors, iodine diet, withdrawal from thyroid
10% to 20% of all thyroid cancers; lymph node metastases are common hormone replacement, or given
and a rare type, Hürthle cell cancer. in recurrence. recombinant human thyroid hormone
• Medullary thyroid cancer (MTC) • Anaplastic cancers are extremely (rhTSH) if there is no evidence of
arises from the parafollicular C cells aggressive, with 5-year survival rates metastatic disease.
and accounts for 1% to 2% of all below 5%. • After surgery and radioactive
thyroid cancers. • The history should include radiation iodine, thyroxine suppression
• Anaplastic thyroid cancer is a rare, exposure, family history, and prevents the growth of microscopic
but rapidly fatal, form of thyroid compressive symptoms (dysphagia, disease.
cancer. hoarseness, pain or pressure) from • External beam radiation is used for
• Other histologic types of cancer, enlarging tumor. persistent, recurrent, anaplastic,
such as lymphoma, sarcoma, and • Concerning examination findings poorly differentiated tumors that are
metastatic cancers, can also be include a fixed mass or not iodine avid.
found within the thyroid. lymphadenopathy. • Chemotherapy is mainly palliative for
• Known risk factors for the • Presented as incidental findings on poorly differentiated or anaplastic
development of thyroid cancer computed tomography (CT), tumors. Traditional chemotherapy
include radiation exposure and magnetic resonance imaging (MRI), has minimal response rates, but
iodine deficiency. positron emission tomography (PET), newer, targeted therapies, such as
• Thyroid cancer can also run in and ultrasound. sorafenib, lenvatinib, and sunitinib,
families or exist as part of familial • Preoperative laboratory studies are showing promise.
syndromes (Gardner, Cowden, and include thyroid-stimulating hormone • Surveillance for recurrent thyroid
Werner syndromes). (TSH) and thyroglobulin (Tg). cancer includes measurements of
• More recently, the molecular • Fine-needle aspiration (FNA) biopsy TSH, Tg, and anti-Tg antibodies in
pathogenesis of thyroid cancer has is a key component of the workup of addition to cervical ultrasound. The
been investigated. The following are thyroid nodules. The Bethesda schedule of these tests is tailored to
the most widely studied molecular criteria classify FNA results and risk level.
markers for DTC to date: determine the risk of cancer in the • Treatment of recurrence can include
○ RET/PTC rearrangement nodule. external beam radiation, targeted
○ BRAF • Preoperative imaging should include therapies, or ultrasound-guided laser
○ PAX8-PPARγ rearrangement cervical ultrasound. CT is used when ablation, depending on the iodine
○ NRAS, KRAS and HRAS aggressive variants are suspected, in avidity of the tumor.

1074
Cancer of the Endocrine System  •  CHAPTER 68 1075

Medullary Thyroid Cancer cardiopulmonary bypass may be Multiple Endocrine Neoplasia


• Medullary thyroid cancer (MTC) necessary for caval involvement. Syndromes
accounts for 1% to 2% of all thyroid • Long-term surveillance, consisting of • MEN1 manifests first with
cancers; 75% of cases are sporadic, physical examinations and CT scans, hyperparathyroidism in patients in
and 25% are familial (multiple is necessary to monitor for disease their 30s and 40s. Other
endocrine neoplasia [MEN] type 2, recurrence. manifestations include pituitary
familial medullary thyroid carcinoma • Mitotane alone or in combination tumors and neuroendocrine tumors
[FMTC]). with other chemotherapeutic agents (NETs) of the pancreas (such as
• The diagnosis is made by FNA with improves recurrence-free survival. gastrinoma) and upper
calcitonin staining and/or washout. • Radiotherapy may improve local gastrointestinal tract.
RET testing can identify inherited control, but there are no clear • MEN1 is inherited in an autosomal
germline mutations. Cervical recommendations. dominant fashion. Mutations in the
ultrasound or CT scans assist with • Hormonal control can also limit MENIN tumor suppressor gene
operative planning. The tumor disease spread and consists of cause this disease, but expression is
markers calcitonin and mitotane, ketoconazole, metyrapone, variable.
carcinoembryonic antigen (CEA) can and etomidate. • Treatment of parathyroid hyperplasia
be useful in following patients • Excision or reoperation is is subtotal parathyroidectomy and
postoperatively for identifying recommended for recurrent or bilateral cervical thymectomy.
recurrence and metastases. metastatic disease. • MEN1 patients with pancreatic and
• At a minimum, treatment of clinical • The prognosis is poor, with an duodenal tumors are treated with
MTC should consist of total overall 5-year survival rate of less distal pancreatectomy, enucleation
thyroidectomy plus central lymph than 40%. of pancreatic head tumors,
node dissection. Lateral neck Malignant Pheochromocytoma duodenotomy, and mucosal
dissection is added when there are • The incidence of malignant resection of multiple duodenal
clinically positive nodes in the pheochromocytoma (PCC) is 2 to 8 tumors.
central neck and for high-risk per 1,000,000 adults. • MEN2A is characterized by
patients. • Most malignant PCCs are sporadic, PCC, MTC, and primary
• Traditional chemotherapy is not but 10% are part of inherited hyperparathyroidism
effective for metastatic MTC, but syndromes such as MEN syndromes, (hyperplasia).
newer, targeted therapies for neurofibromatosis type 1, von • MEN2B is characterized by more
metastatic disease, such as Hippel-Lindau syndrome, and aggressive MTC, PCC, and mucosal
vandetanib and cabozantinib, have succinate dehydrogenase gene ganglioneuromas.
shown some promise. mutations. • The MTC in MEN2 syndromes arises
from C-cell hyperplasia and germline
Adrenocortical Cancer • PCCs manifest with the classic triad
of functional tumors—headache, RET mutations.
• The incidence of adrenocortical
tachycardia, and sweating—but they • Specific codon mutations in the RET
cancer is 1 to 2 per million
are asymptomatic in more than 50% gene determine the disease
people.
of cases, incidentally discovered as phenotype in MEN2 syndromes, and
• Most adrenocortical cancers are
an adrenal mass. help risk-stratify patients.
sporadic, but they can also occur as
• The diagnosis is established with • Prophylactic thyroidectomy should
part of familial syndromes such as
urinary or plasma fractionated be offered to mutation carriers;
MEN1, Li-Fraumeni syndrome,
Beckwith-Wiedemann syndrome, metanephrines and catecholamines. the timing of thyroidectomy is
• Tumors are localized with CT or MRI; determined by the specific codon
and Carney complex.
metaiodobenzylguanidine (MIBG) is mutation.
• Most are asymptomatic, but 40% to
used to identify extraadrenal • PCCs are often bilateral in MEN2
60% are functional (hormone
metastases. syndromes, but onset is
production), and this may be the
• Treatment begins with surgery to asynchronous. Consequently,
presenting symptom(s).
resect the entire gland with clear prophylactic adrenalectomy is not
• The diagnosis is by urinary or
plasma biochemical testing and margins. Surgery can also be useful indicated. Those MEN2 patients who
in debulking for metastatic disease. develop bilateral disease can be
imaging: CT, fluorodeoxyglucose
• Metastatic or unresectable disease treated with bilateral adrenalectomy
(FDG)-PET.
can be treated with 131I-MIBG or and hormone replacement or
• Often the diagnosis is not made
chemotherapy. cortical-sparing adrenalectomy.
definitively until after resection of
suspicious masses, and pathologic • Radiotherapy is used for palliation in Carcinoid Tumors
assessment provides definitive bone and lymph node metastases. • The incidence of carcinoid tumors is
diagnosis. FNA of adrenal masses is • Medical therapy to prepare patients 5.25 per 100,000 people.
rarely indicated. for surgery or control symptoms of • Tumors are identified with specific
• Surgery is the mainstay of treatment catecholamine excess can include immunohistochemical staining for
for adrenocortical cancer and should phenoxybenzamine, nicardipine, or neuron-specific enolase (NSE) or
consist of en bloc resection of the metyrosine. chromogranin A. Chromogranin A
adrenal gland with adjacent organs • Five-year survival ranges from 20% also serves as a blood marker for
and tissue that is involved; to 50%. the disease.
Continued
1076 Part III: Specific Malignancies

• Several classification systems exist • Pancreatic neuroendocrine tumors • Clinical characteristics of parathyroid
for carcinoid tumors, including the (pNETs) can be sporadic or inherited carcinoma include the constitutional
World Health Organization (WHO) (MEN). symptoms of primary
classification and the European • pNETs are diagnosed with CT or hyperparathyroidism, including
Neuroendocrine Tumor Society MRI imaging; ultrasound or muscle weakness, fatigue, nausea,
(ENETS) staging system. endoscopic ultrasound (EUS) vomiting, increased thirst, and
• Carcinoids arise from the Kulchitsky examination can help guide biopsy. frequent urination, in addition to
cells in crypts of Lieberkühn of • The ENETS staging system is bone pain and fractures.
the gut or disseminated in the proposed to help stage pNETs. • A neck mass occurs in 34% to 52%
endobronchial mucosa, and are • Insulinomas are diagnosed with but is uncommon in benign
classified by location. fasting hypoglycemia with elevated parathyroid adenomas.
• The diagnosis is made definitively plasma insulin levels; 10% are • In parathyroid carcinoma, serum
through tissue diagnosis, but urinary malignant, and surgical resection calcium is quite elevated (14.6–15.9)
5-hydroxyindoleacetic acid (5-HIAA), (enucleation) is curative. with elevated serum parathyroid
serum NSE, and chromogranin A are • Glucagonoma is characterized by hormone (PTH) (commonly 10-fold
serum markers of the disease. migratory necrotizing erythema, higher than the upper limit of
• CT or MRI can be used to localize insulin-resistant diabetes, glossitis, normal).
the carcinoid tumors, gallium-68 ileus, and constipation; 50% to 80% • The diagnosis is made by means of
(68Ga)–DOTATATE PET-CT or are metastatic. laboratory measurement of Ca and
OctreoScan can be used, because • Because of the higher rate of PTH, and then technetium-99m
these tumors have somatostatin metastatic disease, surgical sestamibi scan and neck ultrasound
receptors. resection is curative in less than can be used to localize the tumor
• The carcinoid syndrome occurs one-third of patients with with or without washout for PTH
in metastatic carcinoid and glucagonoma. measurement in FNA material.
presents as flushing, diarrhea, and • Somatostatinoma is characterized by • Pathologic features of parathyroid
bronchoconstriction. Right-sided diabetes, diarrhea, and gallbladder carcinoma include local invasion and
valvular heart disease is also a disorders. lymph node metastases.
manifestation of the disease. • Treatment for somatostatinoma • Treatment of parathyroid carcinoma
• Treatment begins with resection of includes cytoreductive surgery and should include en bloc resection of
primary tumor with nodal chemotherapy. the parathyroid mass with ipsilateral
metastases. • Gastrinoma is characterized by ulcer thyroid lobe with or without
• Debulking or metastasectomy is disease, in spite of adequate ipsilateral neck dissection followed
beneficial for controlling symptoms treatment, and diarrhea. by postoperative calcium and
in patients with liver disease or bulky • Gastrinoma is diagnosed through activated vitamin D supplementation.
disease. hypergastrinemia with elevated basal • Medical therapy for hypercalcemia
• Radiation therapy is rarely used acid output or positive secretin test precipitated by parathyroid
for primary therapy but can be result; tumors are localized with CT, carcinoma should start with
palliative. MRI, or octreotide scan. These hydration and loop diuretics;
• Antihormonal therapy consists of tumors are frequently metastatic. calcimimetics (Cinacalcet) or
octapeptide analogues of • Targeted therapies such as bisphosphonates can later be added
somatostatin; Sandostatin LAR is a everolimus and sunitinib have been to lower the serum calcium levels.
helpful, long-acting formulation approved by the US Food and Drug • Adjuvant therapy includes
octapeptide analogue of Administration (FDA) for first-line chemotherapy, such as dacarbazine,
somatostatin. treatment of pNETs. 5-fluorouracil (5-FU), or
• The liver is a common site for Parathyroid Carcinoma cyclophosphamide; radiotherapy is
metastatic carcinoid, and there are • The incidence of parathyroid of limited efficacy.
several options for hepatic-directed carcinoma is 5.73 per 10 million • Patients with features of
therapy, including surgery and people. hyperparathyroidism–jaw tumor
embolization (chemoembolization or • The etiology of parathyroid cancer syndrome or a family history should
radioembolization). has recently been attributed to undergo genetic counseling and
• Metastatic disease can also be pericentromeric inversion resulting in HRPT2 testing.
treated with targeted agents or overexpression of the cyclin D1 • After surgery, one-third of patients
emerging radionuclide therapy. gene. are cured, one-third have recurrence
Pancreatic Neuroendocrine • In hyperparathyroidism–jaw tumor after prolonged disease-free survival,
Tumors syndrome, there are mutations in the and one-third experience a short,
• The incidence of NETs is 2.4% to HRPT2 tumor suppressor gene aggressive course; the 5-year
5.8% per 100,000 people. (parafibromin). survival is 82.5%.
Cancer of the Endocrine System  •  CHAPTER 68 1077

Unlike tumors found elsewhere in the body, cancers of the endocrine


organs can cause symptoms of hormonal excess in addition to mass Table 68.1  Histologic Classification of Thyroid
effect, obstruction, or pain from the mass itself. Therefore physicians Cancers and Their Incidence
who care for patients with endocrine cancers must combat both the
Tumor Histology Incidence (%)
physiologic manifestations and the neoplasia. Any of the endocrine
tumors can be part of a multitude of familial syndromes, so the clinician Differentiated carcinomas 81–87
caring for these patients must always keep this possibility in mind.  Papillary
This chapter covers selected endocrine cancers and highlights some   Follicular variant of papillary
of the unique challenges in treating such tumors. Many common Follicular and Hürthle cell
themes emerge. Diagnosis typically involves biochemical confirmation  Medullary 6–8
of the endocrinopathy followed by imaging to locate the tumor(s).  Anaplastic 5
Surgery plays a role in the initial treatment and often for recurrence  Lymphoma 1–5
and palliation. Traditional chemotherapy is of limited use for endocrine  Metastatic <1
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. 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.
THYROID CANCER Historically, patients received radiation treatments for enlarged tonsils
Incidence or facial acne. Today, patients with cancer such as Hodgkin disease
might still receive radiation treatments. In addition, children exposed
Thyroid cancer is the most common endocrine cancer. A spectrum to radioactive fallout from the Chernobyl (Russia) accident have
of biologic behavior exists, ranging from indolent, well-differentiated demonstrated an increased incidence of thyroid cancer.11,12 Based on
tumors to extremely aggressive, poorly differentiated or anaplastic evidence from patients radiated for Hodgkin disease, doses of 40 Gy
cancers.1,2 Thyroid cancer is the most rapidly increasing malignancy are potentially carcinogenic.13 Epidemiologic studies have reported
in the United States for both men and women. From 1980 to 2006, that 7% to 9% of patients who received 5 to 10 Gy of external beam
the annual US thyroid cancer age-adjusted incidence rose from 4.33 radiation develop thyroid cancer.14 A lag time of 10 to 20 years usually
to 11.03 cases per 100,000 population. This incidence increased by exists between exposure and diagnosis of thyroid cancer, although
2009 to 14.3 per 100,000.3 This is a nearly threefold increase in much shorter periods have been reported (Table 68.2).14
incidence. The gender-adjusted incidence rose from 6.5 to 21.4 = Included in the environmental etiology for thyroid cancer is dietary
14.9 per 100,000 women and almost 4 times greater than that of iodine content. A higher incidence of PTC exists in regions with
men, from 3.1 to 6.9 = 3.8 per 100,000 men.3 This increasing incidence high dietary iodine content, such as the Pacific rim and Iceland.15
is attributed to improved detection of smaller tumors, mostly papillary Iodine-deficient countries, in contrast, experience a higher incidence
thyroid cancer, using high-resolution neck ultrasonography.4 This of FTC in addition to benign thyroid goiters. Many factors con-
change has been attributed mostly to the increase of papillary thyroid found these studies that link changes in DTC rates to iodine intake.
cancer (PTC) incidence.5 A published report from a population-based Ethnicity, selenium, goitrogen, and carcinogen intake likely play
study states that the rapid increase in incidence of thyroid cancer is causative roles.16
attributed to occult cancer detected through neck imaging with stable Increasing investigation into molecular markers that can distinguish
clinically detected thyroid cancer and disease mortality.6 Despite this carcinoma from benign nodules has led to a greater understanding
improved detection, the mortality rate remains unchanged at 0.5 per of the genetic alterations in thyroid cancer. For example, 70% of
100,000 population.7,8 Therefore, thyroid cancer presents a unique cancers found in Chernobyl survivors carried an RET and PTC gene
challenge to the treating physician to manage patient expectations, (RET/PTC) rearrangement. The fusion of the tyrosine kinase encoding
minimize potentially lifelong complications, use the appropriate surveil- domain of the RET protein with a heterologous group of genes occurs
lance for follow-up, and identify patients with more aggressive, poorly in 20% to 40% cases of PTC and is called the RET/PTC rearrangement.17
differentiated forms. RET/PTC rearrangements are frequent in small, multifocal PTCs
accompanied by an inflammatory infiltrate, often seen in individuals
Classification exposed to ionizing radiation and in children.18 BRAF is a member
of the RAF-MEK-ERK serine/threonine kinase-signaling cascade, and
The most common type of thyroid cancer is PTC, representing 80% a BRAF mutation is found in 40% to 60% of PTC cases.19,20 The
of all cases. The second most common type is follicular thyroid cancer V600E BRAF point mutation or mutations in another member of
(FTC), which represents 10% to 20% of all cases. Together, papillary this signaling pathway, RAS, are frequent in cases of poorly differentiated
and follicular cancers are termed differentiated thyroid cancer (DTC), PTC or ATC.21 Analysis of 27 PTCs by Nikiforova and colleagues
and both arise from the thyroid follicular cells. MTC comes from the showed that 70% harbored mutated genes, distributed among BRAF
parafollicular C cells. This neuroendocrine thyroid tumor represents (60%), PIK3A (11%), TP53 (7%), and NRAS (4%). In the same
5% to 10% of all thyroid cancer cases and occurs in familial and study, the prevalence of mutated genes in follicular carcinoma was
sporadic forms. Finally, anaplastic thyroid cancer (ATC) is one of the NRAS, 25%; KRAS, 0.5%; HRAS, 0.2%; TSHR, 11%; TP53, 11%;
most aggressive and rapidly fatal cancers. It can develop from DTC and PTEN, 0.2%.20 Most BRAF substitutions keep the protein in a
that dedifferentiates over time, and it also arises de novo.1,2,9,10 The catalytically active form, resulting in constitutive activation of the
first part of this section on thyroid cancer discusses DTC, and the RAF-MEK-ERK signaling cascade and constant mitogenic activity.
second part reviews MTC (Table 68.1, Fig. 68.1). A very high specificity of approximately 99% of BRAF V600E has
been estimated in data from multiple studies, with very low sensitivity
Etiology to rule out malignancy.22
A higher prevalence of BRAF V600E mutations were observed in
External radiation exposure to the cervical region is one of the most papillary thyroid microcarcinoma with lymph node metastasis and
well-known causes of thyroid cancer. This risk is related to radiation tumor recurrence.23–25
1078 Part III: Specific Malignancies

A B

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

The Ras proteins are plasma membrane guanosine triphosphatases have familiar occurrence.5 Compared with sporadic DTC, FNMTC
activated by growth factor receptors. Mutations that result in their is more aggressive with increased recurrence and decreased disease-free
constitutive activation lead to oncogenesis. RAS mutations occur in survival, local invasion, multicentricity, lymph node metastases, invasion
approximately 40% of follicular cancers and in a small portion of to surrounding structures, and combination with chronic lymphocytic
PTCs, particularly the follicular variant of PTC.26,27 Similar to the thyroiditis.45,46 However, in the absence of a suitable genetic test,
RET/PTC rearrangement, another interchromosomal translocation families cannot be screened, and FNMTC is difficult to distinguish
occurs in FTC. The promoter element of the gene encoding paired from sporadic DTC.47
box 8 (PAX8) fuses with the coding sequence of the peroxisome
proliferator-activated receptor γ (PPARγ) gene in 35% of FTCs.21,28,29 Classification and Prognosis
The functional consequences of the PAX8/PPARγ rearrangement remain
unclear. RAS mutations are also highly prevalent in FTC, but Nikiforova DTCs are divided broadly as papillary or follicular. Follicular variant
and colleagues found that RAS and PAX8/PPARγ rearrangements are PTC has features of both PTC and FTC but is classified as an PTC
mutually exclusive, suggesting that these are two distinct molecular subtype (see Table 68.1 and Fig. 68.1A). In general, well-differentiated
pathways for FTC development.21 PTC has an excellent prognosis, with 5-year survival greater than
For diagnostic accuracy of molecular markers, especially for 97%.7 Smaller tumors carry a better prognosis than larger tumors.
indeterminate thyroid nodules, multiple panels have been expanded PTCs smaller than 1 cm are called papillary microcarcinomas and have
to include mutational/translocational BRAF, NRAS, HRAS, and KRAS been reported in 10% to 30% of autopsy studies.48–50 In the past,
point mutations, in addition to RET/PTC1 and RET/PTC3, with these tumors were incidentally detected in thyroidectomy specimens,
or without PAX8/PPARγ rearrangements.30–34 TERT promoter mutations but they are now detected with increasing frequency with high-
have been identified in 2% to 10% of sporadic PTCs.35 They may resolution ultrasonography. They are believed to have an excellent
coexist with the V600E BRAF mutation in PTC and tend to be more prognosis, with a structural disease recurrence rate of 1% to 2% in
aggressive.36 unifocal PTCs and 4% to 6% in multifocal PTCs.51,52 However, some
Aside from these acquired genetic lesions, some forms of DTC may behave more aggressively than previously appreciated, and manage-
are also inherited. DTC is seen in familial syndromes such as Gardner ment remains controversial.53,54
syndrome, Cowden syndrome, and Werner syndrome.37,38 Familial Age is another important determinant of prognosis in DTC. Older
nonmedullary thyroid cancer (FNMTC), in which two or more patients tend to have more poorly differentiated, aggressive variants
first-degree relatives have been diagnosed with DTC in the absence and are less likely to respond to radioactive iodine (RAI). In these
of another syndrome, exhibits autosomal dominant behavior with cases, death results from local invasion and extensive metastases.
incomplete penetrance and variable expressivity.38–40 Linkage analyses Therefore the completeness of resection and extrathyroidal extension
have identified several candidate genes for FNMTC, including TCO1, are two prognostic indicators used in many staging systems for DTC.55,56
MNG1, fPTC/PRN, and NMTC1, but a single responsible gene has The role of lymph node metastases in determining DTC-specific
not been identified.41–44 Evidence has shown that 5% to 10% of DTCs survival remains controversial. Lymph node involvement is common
Cancer of the Endocrine System  •  CHAPTER 68 1079

Table 68.2  Risk Factors for Malignancy in Nodular Table 68.3  TNM Classification of Malignant
Thyroid Tumors of the Thyroid Gland
LOW RISK ↔ HIGH RISK PRIMARY TUMOR (T STAGE)
Factor 1 2 3 4 5 Tx Tumor cannot be assessed
Age T0 No clinical evidence of tumor
Elderly • T1 Tumor ≤2 cm limited to the thyroid
Child • T2 Tumor >2 cm and <4 cm limited to the thyroid
Sex T3 Tumor ≥4 cm limited to the thyroid or any tumor with
minimal extrathyroid extension
Male •
T4a Tumor of any size extending beyond the thyroid capsule
Female • to invade the subcutaneous soft tissues, larynx, trachea,
Low-dose radiation in childhood • esophagus, or recurrent laryngeal nerve
Family history • T4b Tumor that invades prevertebral fascia or encases
Cystic mass • carotid artery or mediastinal vessels
Solid mass • ANAPLASTIC CARCINOMASa
Multiple masses • T4a Intrathyroidal—surgically resectable
Solitary mass • T4b Extrathyroidal—surgically unresectable
Growing mass •
REGIONAL LYMPH NODES (N STAGE)
Stable mass •
Nx Regional nodes cannot be assessed
Hot scan •
N0 No palpable nodes
Cold scan •
N1 Regional nodal metastases
Warm scan •
N1a Level VI nodes (pretracheal, paratracheal, prelaryngeal)
Fine-needle aspiration (−) •
N1b Metastasis to unilateral, bilateral, or contralateral cervical
Fine-needle aspiration (+) • or superior mediastinal nodes
Associated cervical adenopathy •
DISTANT METASTASES (M STAGE)
Complete resolution in response to •
thyroid suppression Mx Metastases cannot be assessed
Partial resolution in response to • M0 No evidence of distant metastases
thyroid suppression M1 Distant metastases present
No response to suppression •
a
All anaplastic carcinomas are considered T4 tumors.
Modified from Sessions RB, Diehi WL. Thyroid cancer and related nodularity. Modified from Greene FL, Page DL, Fleming ID, et al, eds. AJCC Cancer Staging
In: Myers E, Suen J, es. Cancer of the Head and Neck. 2nd ed. New York: Churchill Manual. 6th ed. New York: Springer-Verlag; 2002.
Livingstone; 1981:766.

in PTC, but the exact incidence of lymph node metastases depends preoperatively, then the risk of nodal recurrence was only 1.5%. Of
on how it is defined. Palpable disease in the lymph nodes is present note, in this study of 590 patients with microcarcinomas, 40% of
in 5% to 10% of patients with PTC, but ultrasound detects pathologi- patients had lateral neck lymph node metastases identified histologically
cally positive lymph nodes in 30% of patients.57,58 Ultrasound features after prophylactic neck dissection.62 Hence, lymph node metastases
that are associated with thyroid cancer include nodule hypoechogenicity do affect recurrence, and clinically apparent nodes are more important
compared with the surrounding structures, microcalcifications, irregular than pathologically positive nodes. The impact of lymph nodes on
margins, and taller-than-wide shape as measured on a transverse view.5 survival is less clear. Large series and population-based studies have
Only 2% of patients with FTC have lymph node metastases because suggested that there is a small but significant effect on survival.63,64
the route of spread is mostly hematogenous, but treatment guidelines From the Surveillance, Epidemiology, and End Results (SEER)
and retrospective studies frequently consider PTC and FTC together. database, comprehensive analysis showed that patients younger than
Routine histologic examination of lymph nodes reveals DTC in 20% 45 years with lymph node metastasis had small but significant risk of
to 50% of patients (particularly papillary carcinoma), but when more death compared with younger patients with no lymph node involve-
detailed inspection is performed, up to 90% of patients with DTC ment.65 Because of the questionable effect on mortality, lymph node
will have lymph nodes with microscopic disease, smaller than 2 mm.59,60 status is not included in all of the staging systems available for DTC.
An increasing amount of high-quality evidence supports sonographic For example, the AGES system considers age, grade, extrathyroidal
survey of cervical lymph nodes in all patients with thyroid nodules.5 extension, and size.66 The AMES system uses age, distant (non–lymph
Historically, lymph node involvement was believed to increase local node) metastases, extent of primary tumor, and size.67 Some, such as
recurrence without affecting survival, and therefore surgeons took a the MACIS system (metastases, age, complete excision, invasion, and
conservative approach to lymph node dissection for DTC. Wada and size), also account for the adequacy of surgical treatment.68 Alternatively,
colleagues demonstrated that patients with pathologically positive staging systems developed by the Ohio State University,69 the European
lymph nodes had a recurrence rate of 16.3% compared with 0% in Organisation for Research and Treatment of Cancer (EORTC),70 the
patients without pathologically positive lymph nodes.61 Whether National Thyroid Cancer Treatment Cooperative Study (NTCTCS),71
metastatic lymph nodes are evident preoperatively appears to be and the American Joint Committee on Cancer (AJCC)72 all do consider
an important factor determining recurrence. For example, Ito and lymph node status. The AJCC staging system is the most widely used
colleagues found that if metastatic lymph nodes were not seen (Table 68.3). It is also known as the TNM system because it considers
1080 Part III: Specific Malignancies

and colleagues: “poorly differentiated thyroid carcinoma is a concept


Table 68.4  Staging of Thyroid Cancer proposed to include carcinomas of follicular thyroid epithelium that
Stage TNM Stage TNM retain sufficient differentiation to produce scattered small follicular
structures and some thyroglobulin (Tg), but generally lack the usual
PATIENTS YOUNGER THAN MEDULLARY THYROID morphologic characteristics of papillary and follicular carcinoma.”82
45 YEARS CANCER These tumors include insular, large cell, tall cell, columnar cell, solid,
I Any T, any N, M0 I T1, N0, M0 and diffuse sclerosing variants.1,82 In patients with these variants, the
II Any T, any N, M1 cancer tends to recur and metastasize. Furthermore, dedifferentiation
II T2–3, N0, M0
of thyroid cancers leads to underexpression or disordered assembly of
PATIENTS AGE 45 YEARS OR III T1–3, N1a, M0 the sodium-iodide symporter, decreasing the usefulness of RAI for
OLDER IVA T4a, N0–1a, M0 treating micrometastatic disease or detection of metastases.83 For these
I T1, N0, M0 T1–4a, N1b, M0 reasons, poorly differentiated thyroid cancers have a 51% disease-free
II T2, N0, M0 IVB T4b, any N, M0 survival rate and a 70% cause-specific survival rate at 5 years.1,81
III T3, N0, M0 IVC Any T, any N, M1 Primary lymphoma of the thyroid is not as common as DTC.
T1–3, N1a, M0 Older women or patients with Hashimoto thyroiditis are at highest
ANAPLASTIC CANCER risk for developing thyroid lymphoma.84 These tumors typically manifest
IVA T4a, N0–1a, M0
IVA T4a, any N, M0 as a rapidly expanding mass causing pain and compressive symptoms.
T1–4a, N1b, M0
IVB T4b, any N, M0 Flow cytometry of cytologic specimens can sometimes be used to
IVB T4b, any N, M0 make the diagnosis, but the condition might be mistaken for Hashimoto
IVC Any T, any N, M1
IVC Any T, any N, M1 thyroiditis. Consequently, a core biopsy is sometimes necessary when
this diagnosis is suspected. Most are B-cell lymphomas treated with
From Greene FL, Page DL, Fleming ID, et al, eds. AJCC Cancer Staging Manual. chemotherapy and radiation. Surgery is occasionally needed for pal-
6th ed. New York: Springer-Verlag; 2002. liation.84 Prognosis depends on the histologic subtype.

Diagnosis
tumor size (T), lymph node metastases (N), and distant metastases Just as with a newly discovered mass anywhere else in the body, the
(M). Like many of the other thyroid cancer staging systems, it also workup of a thyroid nodule begins with a thorough history and physical
considers age, with two different classifications for those younger and examination. A strong family history of thyroid cancer or cancer
older than 45 years. In those younger than 45 years, patients with syndromes or a history of radiation exposure to the head and neck
lymph node metastases are classified as stage I unless they have distant or total-body radiation for bone marrow transplantation85 should raise
metastases (stage II) (Table 68.4).72 the suspicion of thyroid cancer. Rapid growth and/or hoarseness with
It is worth noting some important ways in which FTC differs compressive symptoms may indicate that the thyroid nodule is thyroid
from PTC (see Table 68.1, Fig. 68.1B). Pure follicular carcinoma lymphoma or a poorly differentiated thyroid cancer.1,2,14 On examina-
tends to occur more in older patients, and it carries a worse prognosis tion, malignant nodules are harder and fixed, whereas a nodule that
than PTC. Follicular carcinoma is more common in women than in is rubbery or soft and moves easily with deglutition is reassuring but
men by approximately three times.73 Even when the disease is confined not diagnostic of a benign nodule. Nonpalpable nodules have the
to the thyroid, 5% to 15% of patients ultimately die from the disease, same risk of malignancy as palpable nodules of the same size.86–89
although survival still extends decades as in PTC.74 In addition to the Cervical lymphadenopathy also increases the likelihood that a thyroid
prognostic factors common to the aforementioned DTC staging systems, nodule is malignant.2,90
prognosis in FTC depends on the degree of capsular and vascular
invasion. Minimally invasive tumors are grossly contained within the Laboratory Studies
thyroid but have microscopic foci of invasion into the capsule. Invasive Because the management of patients with functional thyroid nodules
tumors carry a worse prognosis and invade the capsule and vessels.75,76 differs from that of patients with nonfunctional nodules, obtaining
Hürthle cell tumors of the thyroid are often classified with follicular a thyroid-stimulating hormone (TSH) measurement early in the workup
cancer because they are derived from the follicular cell. Both adenomas of a thyroid nodule can efficiently identify patients with a nodule and
and carcinomas of the Hürthle cell can occur, and differentiating hyperthyroidism. In this subset of patients with a suppressed TSH,
them by cytologic assessment is difficult, as it is with follicular an iodine-123 (123I) scan can distinguish a solitary toxic nodule from
lesions.77,78 Capsular and vascular invasion distinguish carcinoma from a toxic multinodular goiter and Graves disease. A solitary hyperfunction-
adenomas. Large Hürthle cell cancers (>2 cm) have a higher recurrence ing nodule is rarely malignant, and fine-needle aspiration (FNA) biopsy
rate, ranging from 21% to 59%.77,79 Furthermore, Hürthle cell cancers or further cancer workup is rarely necessary. The one exception is that
do not always concentrate iodine. For these reasons, Hürthle cell functioning nodules in children do carry a higher risk of malignancy.91
carcinoma carries a worse prognosis compared with DTC. Adenomas If thyroid radionuclide scanning is undertaken, “cold” nodules should
have an excellent prognosis after resection and less than 2.5% dem- undergo FNA biopsy because 10% to 20% of cold nodules are
onstrate malignant behavior, but resection is recommended for larger malignant.92,93 A higher serum TSH is an independent risk factor for
adenomas because size is a major predictor of malignancy.79,80 predicting malignancy and is associated with more advance disease.94,95
As follicular or papillary cancers progress or dedifferentiate, their Other laboratory tests can be helpful once the diagnosis of a certain
prognosis becomes much worse. Anaplastic cancers are at the least type of thyroid cancer has been made. For example, measuring serum
differentiated end of the spectrum and represent one of the most Tg in patients with DTC can assist with the long-term follow-up of
aggressive cancers, with 5-year disease-free survival and cause-specific patients treated for DTC.96–98 Although Tg levels can be elevated in
survival rates of 0%.81 ATC can arise from well-differentiated tumors, patients with DTC, the test is insensitive and nonspecific for diagnosing
or it can also develop de novo (see Fig. 68.1C).81 A group of tumors cancer, elevations in Tg can occur in benign thyroid disorders, and
falls between well-differentiated thyroid cancers and anaplastic cancers. the American Thyroid Association (ATA) guidelines do not recommend
These cancers, called poorly differentiated thyroid cancers, are intermediate routine preoperative Tg measurement for patients with DTC. After
in terms of their histologic appearance and their biologic behavior.1 a total thyroidectomy, however, elevations in Tg can reliably indicate
Although the literature remains inconsistent about what constitutes recurrent or metastatic disease.5 Different threshold Tg levels can
poorly differentiated cancer, the best definition comes from Burman indicate recurrence depending on the concomitant TSH level. It should
Cancer of the Endocrine System  •  CHAPTER 68 1081

be emphasized, however, that there is no role for Tg measurement in Testing for molecular markers from FNA aspirates could be
the initial evaluation of thyroid nodules. conducted for FLUS, AUS, or follicular neoplasm. These are BRAF
Measurement of serum anti–thyroid peroxidase (TPO) antibod- and RAS mutational status, high-density genomic data for molecular
ies may be helpful in patients with high TSH levels suggestive of classification (an FNA-trained mRNA classifier), and FNA-trained
autoimmune thyroiditis; however, routine measurement is not necessary. miRNA classifier combined with molecular markers of malignancy.107,108
Patient counseling regarding the potential benefits and limitations and
Fine-Needle Aspiration Biopsy the long-term clinical implications of the molecular testing should
FNA biopsy remains the gold standard for evaluating thyroid nodules. be done according to the ATA guidelines. The guidelines also recom-
Most clinical practice guidelines recommend FNA biopsy for nodules mend that these tests should be performed in Clinical Laboratory
greater than 1 cm in largest dimension with a high to intermediate Improvement Amendments/College of American Pathology (CLIA/
sonographic pattern.5,92,99 Biopsy can be performed on nodules larger CAP)–certified molecular laboratories or the international equivalent.5
than 1.5 cm with few suspicious characteristics.5 For nodules larger
than 1 cm with suspicious sonographic features of extrathyroidal Imaging
extension or associated lymphadenopathy, FNA should be performed Not only can ultrasonography improve the accuracy of FNA biopsy,
in selected patients such as those with family history of thyroid cancer.100 but it is also an important tool in evaluation of thyroid nodules
When the FNA result is clearly benign or malignant, then the decision because it is used to measure the size and features of the nodule and
for further treatment, including thyroidectomy, becomes evident. The it can also reveal additional nonpalpable nodules. Ultrasound examina-
false-negative rate for FNA biopsy is 1% to 3% (Table 68.5). The tion alone can increase the clinician’s suspicion for malignancy if the
false-negative rate increases to 10% to 15% when the nodule is large nodule has fine microcalcifications, irregular borders, chaotic vascular
(>4 cm).99,101 Other clinical scenarios in which the clinician should patterns or peripheral vascularity, cystic aspect, or hyperechogenicity.109
not always trust a benign FNA result include patients with a family In addition, ultrasound scanning can be used to evaluate the lymph
history of thyroid cancer, patients with a history of radiation exposure, nodes in both the central and the lateral neck compartments, which
and cystic nodules.102 Ultrasound guidance can improve the accuracy may prompt additional FNA biopsy of suspicious lymph nodes or
of FNA biopsy by confirming that the nodule is actually being sampled result in alterations in the surgical plan. Although ultrasound examina-
and by enabling targeting of the most suspicious portions of the tion is highly operator dependent, it is noninvasive and does not
nodule (e.g., the wall of a cyst). This is especially true for nondiagnostic involve any radiation or contrast agent risk to the patient. High-
cytologic findings (>25%–50% cystic component) or nonpalpable or resolution ultrasonography can also demonstrate extracapsular invasion
posteriorly located nodules.103,104 and subtle lymph node involvement.104,110–115 Consequently, ultraso-
Nodules that do not meet FNA criteria, such as suspicious sub- nography is the preferred method to evaluate the thyroid and cervical
centimeter nodules in individuals older than 60 years, single nodules lymph nodes. Although routine screening is not recommended for
with well-defined margins, and nodules with a rim of normal thyroid all patients, those with a strong family history or radiation exposure
parenchyma that is larger than 2 mm, can be observed.100 can undergo ultrasound screening for thyroid nodules. Use of positron
FNA results are classified according to the Bethesda criteria, which emission tomography (PET) and/or computed tomography (CT) scan
indicate the risk of malignancy (see Table 68.5). One of the limitations is helpful for identifying lung or bone tumors in patients at risk for
of cytologic evaluation of thyroid nodules is that it cannot distinguish metastases.112,116
between adenoma and carcinoma in follicular lesions.92,101 Therefore Highly aggressive cancers that may invade local structures, extend
lobectomy with permanent histology may be the best way to make a into the chest, or demonstrate poorly differentiated cytologic features
definitive diagnosis in follicular or indeterminate lesions. Furthermore, require careful preoperative planning. In such cases, CT becomes a
20% to 30% of FNA results fall into the category of indeterminate helpful preoperative imaging study in planning en bloc resection of
cytologic findings, such as atypia of undetermined significance (AUS) other organs aside from the thyroid, understanding the extent of
or follicular lesion of undetermined significance (FLUS).105 Currently, vascular involvement, determining if a thoracic incision is necessary,
many centers have turned to molecular analysis of FNA specimens and planning for reconstruction.1,112,116,117
to help distinguish follicular lesions. Cytologic specimens are analyzed Chest CT or PET-CT imaging studies performed for other medical
for a panel of mutations, including BRAF, RAS, RET/PTC, and PAX8- indications often reveal thyroid nodules incidentally. PET scans
PPARγ rearrangements. The seven-gene mutational panel, most useful demonstrate thyroid masses during the workup and staging of other
when surgery is favored, has sensitivity that ranges from 44% to cancers. This subset of incidentally discovered thyroid nodules deserves
100%.32–34 Although this is an exciting area of research, the clinical special attention because up to 50% of fluorodeoxyglucose (FDG)-avid
usefulness of the various gene panels has varied, and more prospective thyroid nodules will contain thyroid cancer. Therefore, PET-positive
data are needed.19,21,28,81,106 thyroid nodules should undergo FNA biopsy.

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 5–15 Repeat FNAa
lesion of undetermined significance (FLUS)
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

a
Lobectomy also can be considered depending on clinical or sonographic characteristics.
FNA, Fine-needle aspiration.
1082 Part III: Specific Malignancies

Treatment versus 9%, P = .02) compared with less extensive resections.134 Owing
to concerns regarding the accuracy of risk stratification and complica-
Treatment of DTC involves a surgeon, an endocrinologist, a nuclear tions in these retrospective studies, current guidelines recommend
medicine specialist, and, occasionally, a radiation oncologist. A lobectomy for small (<4 cm), unifocal, well-differentiated tumors with
multidisciplinary approach best serves patients with DTC. no lymph node metastases or extrathyroidal extension.5
Another hotly debated topic related to the extent of initial surgery
Surgery for DTC is the role of prophylactic central neck dissection. The most
The extent of surgery for DTC remains controversial. This is especially recent guidelines are consistent with the 2009 guidelines, which state
true for small, encapsulated, well-differentiated tumors, and tumors that “prophylactic central neck dissection may be performed, especially
smaller than 1 cm (microcarcinomas). These are discussed further in patients with advanced primary tumors (T3 or T4) or clinically
later, but for most DTCs 4 cm or larger that were diagnosed preop- involved lymph nodes” and “total thyroidectomy without prophylactic
eratively, most clinicians recommend a total thyroidectomy.5 Controversy central neck dissection may be appropriate for small (T1 or T2),
still exists for lesions larger than 1 cm and smaller than 4 cm with noninvasive, clinically node-negative patients.”5,97
low- or intermediate-risk features. Contrary to the previous guideline The central neck lymph nodes are also classified as level VI lymph
recommendations,118 the new guidelines5 recommend either total nodes and include the paratracheal, perithyroidal, and precricoid lymph
thyroidectomy or lobectomy for treatment. Thyroid lobectomy may nodes. These nodes are found along and behind the recurrent laryngeal
be sufficient for this type of lesion without extrathyroidal extension node, and frequently surround the lower parathyroid gland. Although
and with no evidence of lymph node metastasis. This is based on the level VI lymph nodes contain macroscopic disease in 10% of
evidence from properly selected patients that showed similar clinical cases, when they are removed prophylactically, 32% to 69% of patients
outcomes with either surgical plan.119–123 Furthermore, lobectomy can will have microscopic metastases.135–137
obviate the need for exogenous thyroid hormone. Finally, because the Proponents of prophylactic central neck dissection argue that the
current practice for follow-up depends more on ultrasound findings initial operation is the safest time to remove central neck lymph nodes
and Tg measurement than on whole-body RAI, total thyroidectomy to prevent local recurrences and the complications associated with
is no longer needed to justify postoperative RAI.5 Analysis of 5432 reoperative surgery in the central neck. Wada and colleagues found
patients with PTC from the SEER database found no difference in the recurrence rate in patients treated with therapeutic lymph node
10-year overall survival between total thyroidectomy (4612 patients) dissection to be 21%, whereas patients who underwent prophylactic
and thyroid lobectomy (820 patients).122 Results from two other neck dissection experienced a recurrence rate of only 0.43%. Important
single-center studies showed excellent survival in properly selected to note, those patients without clinically overt nodal disease who did
patients who underwent lobectomy.119,123 Without evidence of high-risk not undergo prophylactic central neck dissection also experienced a
features, initial lobectomy of 1- to 4-cm thyroid carcinomas is more very low recurrence rate of 0.65%. Hence, the absolute differences
cost-effective after 3 years of follow-up.124 On the other hand, the in recurrence are minuscule.61 Several other studies also have supported
rationale for total thyroidectomy is based on tumor biology and current the concept that microscopically positive lymph nodes rarely progress
treatment modalities. DTC, especially PTC, tends to be multicentric, to recurrence, especially after postoperative RAI ablation.138–140 Clinically
with up to 80% of patients having multiple tumor foci and 60% evident lymph node metastases place patients at higher risk for recur-
having bilateral disease when a thorough pathologic examination of rence, and these patients clearly benefit from therapeutic lymph node
the contralateral lobe was performed.10,53,112 A study showed that 43% dissection. Prophylactic central neck dissection reduces an already low
of patients who underwent thyroidectomy for 1- to 4-cm thyroid recurrence rate and potentially eliminates or reduces the need for RAI
cancers had high-risk characteristics that would have necessitated but is also associated with risks such as hypoparathyroidism. The
complete thyroidectomy if lobectomy had been used as initial treat- risk-to-benefit ratio may favor prophylactic central neck dissection in
ment.125 A total thyroidectomy as the initial procedure obviates the a subset of patients, but the putative risk factors that define such a
need for reoperative surgery to remove the contralateral lobe should subset remain unknown.112,141,142
a recurrence become detected. Second, experienced thyroid surgeons Evidence supports a selective approach of prophylactic level VI
can safely perform a total thyroidectomy, with permanent complications node dissections in patients with clinically or radiographically involved
such as recurrent laryngeal nerve injury and hypoparathyroidism lymph nodes or intraoperative detection of metastatic lymph nodes
occurring at a rate of less than 2%.54,117 RAI therapy for ablation of (cN1).143–145 Thyroidectomy begins with proper patient positioning
microscopic disease becomes most effective when the thyroid remnant in the semirecumbent position with the neck extended (Fig. 68.2).
is small or absent. Tg measurement and radioiodine whole-body A transverse, curvilinear incision is made in a suitable skin crease at
scanning are highly sensitive modalities for detection of recurrent or or beneath the level of the cricoid cartilage. Traditionally a Kocher
metastatic disease, but these two methods are most effective when no collar incision was used, but this requires a very large dissection
thyroid tissue remains in the neck.2,126 superiorly to reach the upper pole of the thyroid, placing the patient
Most low-risk cancers carry an excellent prognosis regardless of at risk for postoperative seroma. Intraoperative ultrasound guidance
the extent of thyroidectomy, and there are no randomized prospective can help in assessing the upper extent of the gland and placing the
trials comparing total thyroidectomy and thyroid lobectomy in this incision appropriately. Superior and inferior subplatysmal flaps are
group of patients. In addition, radioiodine may have limited usefulness raised to create a working space around the thyroid. The strap muscles
in low-risk patients.10,53 For these reasons, some researchers favor thyroid are divided in the median raphe and are retracted laterally. It is rarely
lobectomy in low-risk patients. For example, Shaha and colleagues necessary to transect the strap muscles, but this can be accomplished
have reported 20-year follow-up findings in 465 patients with low-risk if the tumor is large or adherent to the overlying muscles. The peri-
DTC. Although the lobectomy group had more local recurrence thyroidal soft tissue is swept off the gland bluntly to identify the
compared with the total thyroidectomy group (4% versus 1%), there boundaries of the thyroid. Because the thyroid is most fixed at the
was no statistical significance.127 Similarly, other groups have also upper pole, these vessels are divided first. Much of this can be
failed to demonstrate any significant effect on survival.128–130 In contrast, accomplished by using energy devices such as the Harmonic scalpel
large retrospective series have demonstrated improvement in recurrence or LigaSure, but in larger vessels clips and/or ties may be required.
rates for total thyroidectomy compared with less extensive opera- The upper pole vessels should be ligated close to the thyroid capsule
tions.69,131–133 In a frequently cited study, Mazzaferri and colleagues to avoid injuring the external branch of the superior laryngeal nerve.
reported on 1355 patients with a mean follow-up of 15.7 years. Patients In addition, the surgeon should remain vigilant for the upper para-
treated with total thyroidectomy experienced significant improvements thyroid gland, which is frequently located near the upper pole vessels.
in recurrence rate (26% versus 40%, P < .02) and mortality rate (6% Next, the thyroid gland is reflected medially. To accomplish this, the
Cancer of the Endocrine System  •  CHAPTER 68 1083

Sternohyoid Superior thyroid


muscle vessels

Thyroid
Internal
cartilage
jugular
vein

Recurrent
laryngeal
Incision Vagus nerve
nerve Inferior
Sternocleidomastoid Common thyroid
muscle carotid artery artery
A B C

D E
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.

middle thyroid vein is ligated. In addition, dividing the thyroid isthmus pockets. Each pocket should be marked with permanent suture so
can also facilitate medial rotation, assuming that the tumor is not that it can easily be found in a reoperative setting.
located within the isthmus. Before any structures along the medial Preoperative FNA or intraoperative frozen section can be used to
border of the gland are divided, the recurrent laryngeal nerve must confirm that enlarged lymph nodes seen on ultrasound harbor metastatic
be identified and its course dissected. The nerve is found medial to disease. Cytologic or pathologic confirmation of lymph node metastases
the upper parathyroid gland and lateral to the lower parathyroid. The should prompt the surgeon to perform a compartment-oriented lymph
parathyroid glands must also be identified and dissected free from the node dissection. Lymph node sampling or “berry-picking” should be
thyroid on an intact vascular pedicle. Once the recurrent laryngeal avoided, because this leaves behind lymph nodes that likely contain
nerve has been identified, the branches of the inferior thyroid artery microscopic disease, which then become more difficult to excise in a
can be divided along the thyroid capsule. The inferior pole also is reoperative setting.
mobilized with a combination of blunt dissection and ligation of the Although “skip” metastases directly to the lateral compartment
inferior thyroid artery. The thyroid is then dissected off the anterior can occur in PTC, the central neck nodes (level VI) are usually the
surface of the trachea with electrocautery or other energy devices to first nodes to receive drainage from the thyroid (Fig. 68.3). The
divide the small vessels contained within the ligament of Berry. Perform- boundaries of the central neck are the carotid sheathes laterally, the
ing the identical procedure on the contralateral lobe completes a total hyoid bone superiorly, and the innominate artery inferiorly.146
thyroidectomy. Lymphadenectomy in this area requires skeletonizing the recurrent
Before passing the specimen off the field, the surgeon should examine laryngeal nerve along its entire cervical course, and removing all the
it to make sure that there is no parathyroid tissue adherent to the fibrofatty tissue along the trachea. Frequently, the lower parathyroid
gland. Any inadvertently removed parathyroid tissue can be finely is invested in this tissue and becomes devascularized with this
minced and reimplanted into either the sternocleidomastoid or the dissection.141
strap muscles. Frozen section of a biopsy specimen of this tissue can A lateral neck dissection usually involves dissection of levels II,
be used to distinguish among fat, parathyroid, or lymph node; this III, and IV (see Fig. 68.3). This dissection puts the spinal accessory,
will also help in avoiding autotransplanting cancer-bearing lymph phrenic, vagus, cervical sensory, sympathetic trunk, hypoglossal, greater
nodes back into the patient. Two or three pockets are created within auricular, and marginal mandibular branch of the facial nerves at
the muscle, and the minced parathyroid tissue is divided among these risk. The extent of node dissection should be guided by preoperative
1084 Part III: Specific Malignancies

and intraoperative ultrasound findings. Usually the great vessels can


be preserved, but more aggressive tumors can invade the internal Radioactive Iodine
jugular vein, and it should be sacrificed in this scenario. In addition, Remnant ablation with RAI is the standard adjuvant treatment for
to nerve injury, chyle leak is another complication of lateral neck selected patients with DTC. Because the primary goal of RAI is remnant
dissection.112,117 ablation, it can be administered only after a total or near-total thy-
roidectomy; 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.5 RAI
is administered 1 to 3 months postoperatively as iodine-131 (131I) 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
IIB 100 mCi for patients with low-risk tumors and higher doses
IB (100–200 mCi) for patients with residual disease, suspected microscopic
IA disease, or more aggressive histologic subtypes (i.e., tall cell, columnar
cell, or insular variants).5,147–149 To stimulate intracellular uptake of
IIA 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
III thyroid hormone replacement over 4 to 6 weeks.2,150 A newer method
VA is to administer recombinant human thyroid-stimulating hormone
VI
(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
IV period of hypothyroidism as with hormone withdrawal. However,
VB
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
VII morbidity.5 The US Food and Drug Administration (FDA) approved
rhTSH for thyroid remnant ablation in patients who do not have
evidence of metastatic disease.151,152 In addition to increasing the TSH
Figure 68.3  •  The central neck nodes (level VI) are usually the first nodes level, clinicians should also prepare patients by instructing them to
to receive drainage from the thyroid. follow a low-iodine diet for 1 to 2 weeks before RAI treatment. This

A B C
Figure 68.4  •  (A) Whole-body scan acquired 24 hours after administration of 2 mCi of iodine-123 (123I). (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 123I. There is intense uptake in the region, but the resolution is not as good as with 123I. There is faint uptake in the liver on the posttreatment
scan as a result of metabolism of radioiodinated thyroid hormones at that site.
Cancer of the Endocrine System  •  CHAPTER 68 1085

least 6 months after treatment. Similarly, men should avoid conception


for at least 6 months after treatment.149,156,166,167 Although a study has
shown that BRAF V600E mutations significantly reduce sodium-iodine
symporter expression and RAI uptake,168 there is no clear role of
molecular testing in guiding postoperative RAI.5,169
Thyroxine Suppression
Because all cells of follicular origin depend on TSH for growth, TSH
suppression through the administration of supraphysiologic doses of
levothyroxine (T4) remains an important strategy for maintaining
disease-free survival and overall survival.170 For high-risk patients
with incomplete resection, tumor invasion into adjacent structures,
or distant metastases, the physician should initially titrate T4 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).5,147 Once patients remain disease-free for at least 2
years, their TSH suppression can be liberalized to within the refer-
ence 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
Figure 68.5  •  Positron emission tomography (PET) scan of a 75-year-old be carefully considered, particularly in older patients. Because of the
woman with anaplastic cancer of the thyroid. Images were acquired 1 hour risk of bone loss, the NCCN guidelines recommend daily calcium
after intravenous injection of 15 mCi fluorine-18 fluorodeoxyglucose (FDG). and vitamin D supplementation for patients on TSH suppression.147
There is intense uptake of FDG in the undifferentiated cancer.
External Beam Radiation
Although 131I is the preferred adjuvant therapy for thyroid carcinoma,
diet requires patients to avoid foods that contain iodized salt, dairy external beam radiation sometimes plays a role in treating this disease.
products, eggs, seafood, soybeans or soy-containing products, and Persistent, recurrent, anaplastic, or poorly differentiated tumors may fail
foods colored with red dye No. 3.148,149 to take up 131I. Treatment of anaplastic thyroid tumors almost always
Although some studies have shown no benefit to RAI therapy,153,154 includes external beam radiation because these tumors often cannot
other studies have demonstrated a reduction in locoregional recurrences be completely resected and do not concentrate iodine. Although no
and distant metastases.69,131 As with the controversy over the extent improvement in overall survival has ever been documented, external
of thyroidectomy, the benefit of RAI for low-risk patients remains beam radiation is often given after resection of poorly differentiated
unclear.150 The most recent ATA (2016) guidelines recommend remnant tumors to reduce the risk of local relapse.171 The group at Memorial
ablation for all but the lowest-risk patients (unifocal, well-differentiated Sloan Kettering Cancer Center has found that up to 85% of poorly dif-
tumor, <1 cm in size, confined to the thyroid gland without lymph ferentiated tumors display some iodine avidity, and therefore treatment
node metastases or multifocal papillary carcinoma with no other adverse with RAI may remain worthwhile. Patients with incompletely resected
findings).5 These recommendations are supported by multiple systematic tumors, unresectable disease, and locoregional recurrence in a previously
reviews including one that was published in 2015.155–157 In these operated field may benefit from external beam radiation.1,171,172
low-risk patients, most of the available evidence suggests that RAI
does not improve disease-specific or disease-free survival.158–161 The Chemotherapy
National Comprehensive Cancer Network (NCCN) guidelines require Because RAI often can be effective treatment for well-differentiated
a more thorough evaluation for the extent of remaining disease after tumors that have metastasized, cytotoxic chemotherapy has not been
thyroidectomy, with a radioiodine scan 1 to 12 weeks postoperatively. extensively evaluated for metastatic thyroid cancers. For large burdens
RAI ablation is not recommended if the stimulated Tg is less than of disease, anaplastic cancers, or poorly differentiated tumors that are
1 ng/mL and the radioiodine scan result is negative.147 not iodine avid, chemotherapy becomes an important treatment
Some studies have shown an increase in the risk of development component after surgery or if the tumor is not resectable. In these
of secondary malignancies after RAI therapy. This has been examined situations, chemotherapy confers minimal effects because these tumors
with use of the National Cancer Institute’s SEER database. Brown carry a very poor prognosis. Historically, doxorubicin was the most
and colleagues found that patients treated for DTC had significantly effective single agent. Combination therapy with doxorubicin and
higher rates of nonthyroid second primary malignancies than expected cisplatin resulted in modest objective response rates.173,174 Newer,
in the general population. Although the excess risk was relatively targeted therapies have shown some promise. Small-molecule tyrosine
small, it was greater in the subset of patients who were treated with kinase inhibitors (such as sorafenib or sunitinib) and antibodies
RAI.162 Iyer and colleagues specifically examined low-risk patients (anti–vascular endothelial growth factor [VEGF]) should be considered
(T1N0) treated with RAI and found that their excess absolute risk in the context of ongoing clinical trials.147,175–177 Sorafenib and lenvatinib
was 4.6 excess cases per 10,000 person-years at risk.163 As discussed have been approved for use in the United States for patients with
earlier, RAI clearly benefits patients with larger tumors and metastatic advanced RAI-refractory DTC. Although there was no improvement
disease, but the increased risk of secondary malignancies in low-risk in overall survival with sorafenib and lenvatinib, both were associated
patients in whom the long-term benefit of RAI is questionable means with prolongation of progression-free survival (PFS) by 5 months.178,179
that careful patient selection for RAI treatment is necessary.
Hematologic malignancies are the most common secondary Recurrence
malignancies after RAI, but there is also an association with kidney,
breast, bladder, skin, and salivary gland cancers.164–166 The more For most DTCs, the long-term survival rate exceeds 95%. Even
commonly noted side effects after radioiodine treatment include dry though disease-specific mortality rates remain quite low (<1%),
mouth, mouth pain, salivary gland swelling (sialadenitis), altered smell recurrence rates exceed mortality rates. For low-risk tumors, the
and taste, conjunctivitis, and fatigue. Women should not be pregnant locoregional recurrence rates range from 2% to 10%180 and distant
at the time of treatment, nor should they become pregnant for at recurrence rates are 1% to 2%.51,181 Higher-risk tumors (larger size,
1086 Part III: Specific Malignancies

extrathyroidal extension, cervical lymph node metastases) carry recur- care.177 A multidisciplinary team experienced with metastatic thyroid
rence rates of 21% to 68%.62,180 For these reasons, a tailored approach cancers best handles these decisions.
to follow-up and treatment of recurrent disease best serves patients
with DTC. Medullary Thyroid Cancer
Surveillance The prevalence of MTC has decreased from previously reported rates
The original tumor characteristics and operative findings dictate the of 5% to 10% of all thyroid cancers to 1% to 2%. This is because
follow-up schedule for DTC. Surveillance consists of measuring serum of the increased incidence of PTC.190 Unlike DTC, MTC arises from
Tg, TSH, and anti-Tg antibodies in addition to imaging. Cervical the parafollicular C cells instead of the follicular epithelium (see Fig.
ultrasound is a highly sensitive test for detection of metastatic 68.1D). Hence, MTC is a neuroendocrine tumor (NET), and it shares
lymphadenopathy. Elevations in Tg in the absence of cervical disease some properties common among neuroendocrine cancers, including
seen on ultrasound suggest distant metastases.5,56 Whole-body radio- secretion of peptide hormones such as calcitonin, serotonin, or vasoac-
iodine scanning is sensitive for detecting iodine-avid bone or pulmonary tive intestinal peptide (VIP). Most cases of MTC are sporadic, but
metastases.96,99 Poorly differentiated, aggressive tumors will not 25% are a result of germline genetic mutations. Hereditary cases occur
concentrate iodine but can be detected with CT scan or FDG-PET either in isolation (familial medullary thyroid carcinoma [FMTC])
(see Fig. 68.5).83 or as part of MEN syndrome type 2 (MEN2A or MEN2B).126,191
The frequency of surveillance depends on the original tumor
characteristics and the AJCC stage.72 For lower-risk tumors, physical Diagnosis
examination, cervical ultrasound, and measurement of TSH, Tg, and Although any thyroid nodule could potentially harbor MTC, historical
anti-Tg antibodies should be performed every 12 months. These studies features that may alert the physician to the potential for MTC include
can be scheduled 3 to 6 months after the initial RAI treatment in a family history of MTC, PCC, hyperparathyroidism, or other mani-
patients with high-risk tumors.5,147,177 festations of MEN2 syndromes.9 The median age of diagnosis is
approximately 50 years.192 In a SEER study that included 1252 patients
Treatment of Recurrent Disease between 1973 and 2002, the authors found that 87% of the cohort
Recurrence in the neck or cervical lymph nodes is best treated surgically. were white and 60% were female.192 As in evaluating all thyroid nodules,
The decision-making process of surgical resection should be based on neck ultrasound examination and FNA play a major role in diagnosing
clinically apparent, macroscopic nodal disease confirmed with neck MTC. In one retrospective study, ultrasound was found to be falsely
ultrasound or CT scanning, rather than depending on Tg elevation negative for ipsilateral and central node involvement in 17% and
alone.143,182,183 Furthermore, small nodal recurrence (<8 mm in the 14%, respectively, of patients with MTC with no ultrasound pathog-
central neck or <10 mm in the lateral neck) can be managed with nomonic features for MTC.193 Hereditary cases are often detected
active surveillance.182,184–186 through genetic screening to identify germline mutations in the RET
Other treatment options such as percutaneous ethanol injection gene. Almost all sporadic cases manifest with a palpable neck mass,
(PEI) have shown some success in retrospective studies limited by which could be either the thyroid mass or a metastatic lymph node.
small sample sizes.187,188 External beam radiation should be considered In contrast to sporadic disease, hereditary disease usually manifests as
for recurrent disease that is unresectable or not iodine avid (Fig. 68.6). multicentric and bilateral and involves the upper part of the thyroid.194,195
Percutaneous ultrasound-guided laser ablation has shown promising However, bilateral disease in sporadic MTC with negative RET
results for metastatic nodal disease.189 RAI cannot ablate bulky nodal mutation can occur in up to 9% of patients.196–198 Lymph node
disease. After surgical resection, radioiodine can effectively ablate metastases occur in 35% to 50% of patients at initial diagnosis.199
micrometastatic disease throughout the body. Therefore, ultrasound evaluation of the central and lateral neck
Medical treatment for recurrent or metastatic disease consists of compartments for suspicious lymph nodes becomes a crucial component
maintaining TSH suppression. Depending on the location of recurrence, to the initial diagnosis.200
health of the patient, tumor risk stratification, and patient preference, Because the parafollicular C cells are concentrated in the upper,
patients with metastatic disease can be referred for experimental posterior portion of each thyroid lobe, many MTCs arise in a posterior
protocols using targeted therapies, can undergo traditional cytotoxic location, causing symptoms such as hoarseness or dysphagia as a result
chemotherapy, or can be treated with watchful waiting and supportive of compression of local structures. If there is any concern for vocal

TARGET

95%IDL

60%IDL

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.
Cancer of the Endocrine System  •  CHAPTER 68 1087

cord function, then direct laryngoscopy should be performed preop- Commercial testing is performed through polymerase chain reaction
eratively.199 Markedly elevated calcitonin levels can cause symptoms (PCR) amplification of the patient’s germline DNA obtained from
such as flushing, diarrhea, and weight loss.201 Distant metastasis to the patient’s white blood cells. A spectrum of tumor aggressiveness
the liver is the most frequent site.202 CT scan is a very sensitive imaging exists among the various RET mutations, and the timing of prophylactic
modality for detection of local nodal and distant lung metastases. thyroidectomy is based on the specific mutation. Once a patient tests
Liver metastases are best detected with MRI, and routine use of positive for a germline RET mutation, the patient should be carefully
fluorine-18 fluorodeoxyglucose (18F-FDG) PET-CT is not recommended counseled regarding the risk to other family members and the patient’s
in MTC evaluation because it is less sensitive.190,203 children. At-risk family members should be identified and also tested
FNA sensitivity in detecting MTC ranges from 50% to 80% and so that prophylactic thyroidectomy can be offered at the appropriate
can be improved with the addition of immunohistochemical staining time. Although some overlap exists for genetic mutations associated
of calcitonin.204,205 A meta-analysis of 15 publications showed an with MEN2A and familial MTC, distinct mutations are usually
accuracy of FNA of less than 50%.206 FNA characteristics of MTC associated with MEN2B.219,220
include the presence of stromal amyloid without thyroid follicles.
Because spindle-shaped cells may be seen, MTC can be mistaken for Treatment
parathyroid carcinoma or ATC unless the specimen is stained for Complete surgical excision is the treatment of choice for MTC. The
calcitonin, chromogranin A (CgA), or carcinoembryonic antigen minimum extent of surgery for patients with clinically apparent disease
(CEA)—substances produced by MTC that confirm the diagnosis. A is a total thyroidectomy with bilateral central neck dissection. Eighty-one
more sensitive technique than immunohistochemistry on cytology percent of patients with palpable disease have central neck lymph
specimens is to measure the calcitonin level in the washout fluid from node metastases, and the addition of central neck dissection improves
an FNA.207 In addition, the presence of calcitonin messenger RNA cure rates over total thyroidectomy alone in patients with clinically
(mRNA) has been performed when the cytologic or histologic diagnosis evident disease at presentation.221,222 The involvement of ipsilateral
remains unclear.208 and/or contralateral nodes has been found to be positively correlated
Several serum markers can confirm the diagnosis of MTC and are with presence and number of central nodes metastasis.223 Furthermore,
useful in following patients for recurrence and metastases. Calcitonin calcitonin level is helpful in preoperative risk assessment of nodal
is commonly elevated in patients with MTC. Although a small percent- disease. No risk of lymph node metastasis was found when serum
age of normal patients will have some elevation in calcitonin, patients calcitonin level was less 20 pg/mL in a study of 300 patients with
with a diagnosis of MTC typically exhibit levels above 100 pg/mL. MTC.224 In this study, metastasis to the ipsilateral central neck,
In borderline cases, the diagnosis can be clarified by stimulating the ipsilateral lateral neck, contralateral central neck, contralateral lateral
calcitonin with either intravenous calcium gluconate or pentagastrin. neck, and upper mediastinum were associated with calcitonin levels
Of note, calcitonin secreted by nonthyroid tissue such as lung cancer greater than 20, 50, 200, and 500 pg/mL, respectively.224 The initial
or prostate cancer does not elevate with calcium gluconate or penta- approach to lateral neck lymph nodes continues to evolve. Historically,
gastrin stimulation. Currently available immunochemiluminometric the initial surgical treatment included an ipsilateral lateral compartment
assays (ICMAs) are more sensitive and specific for monometric calcitonin neck dissection because up to 80% of patients will have ipsilateral
and largely eliminate cross-reactivity with procalcitonin.190 nodal metastases.196 However, current guidelines recommend performing
Before the advent of genetic testing, these stimulated measure- an ipsilateral lateral neck dissection if ultrasound or physical examination
ments were used to screen patients at high risk for MTC.209 The detects lymphadenopathy in the lateral neck, if central compartment
degree of calcitonin elevation correlates with tumor burden, lymph nodes are involved, or if the primary tumor is greater than
with nodal metastases found with basal calcitonin levels of 10 to 1 cm.200 Intraoperative assessment of nodal disease by surgeons has
40 pg/mL and distant metastases found with calcitonin levels greater sensitivity of 64% and specificity of 75%.221 Contralateral lateral neck
than 150 pg/mL. Patients with calcitonin levels greater than 3000 pg/ dissection is added when patients have bilateral tumors or there is
mL are likely to have widely metastatic disease and are unlikely to extensive lymph node disease on the ipsilateral side. Because some
experience a cure.210 patients require extensive neck dissection, these procedures are often
Preoperative measurement of serum CEA can also help risk-stratify staged.199,200 Unlike DTC, in which micrometastatic disease can be
patients. Overall, CEA elevations occur in more than 50% of patients effectively treated with RAI ablation, the only effective treatment for
with MTC, but a preoperative serum CEA level greater than 30 ng/mL MTC is complete surgical resection. Therefore all evident disease must
highly predicts the inability to cure the patient with surgery.211 CEA be resected for the best chance of long-term cure. According to the
levels above 100 ng/mL may signify extensive lymph node and distant revised ATA guidelines, complete thyroidectomy is not indicated unless
metastases. Following CEA levels postoperatively can also be used to the patient has a RET germline mutation, elevation of serum calcitonin
monitor disease progression. Simultaneous increases in CEA level level, or evidence of residual MTC on images.190
and calcitonin level indicate disease progression,190 whereas elevation Prophylactic thyroidectomy is recommended for at-risk family
of CEA level in the presence of a stable calcitonin level is associated members in hereditary MTC. Current recommendations for the timing
with a worse prognosis because it may indicate tumor dedifferentiation of prophylactic thyroidectomy balance the need to remove the at-risk
and distant metastases. However, normal CEA or calcitonin level organ before it develops clinically apparent disease with the risks of
is rare and could represent an advanced dedifferentiated MTC or surgery. In hereditary MTC, an age-related progression exists from
misdiagnosis.190 Other markers, such as CgA and serotonin, may be C-cell hyperplasia to carcinoma, and, ultimately, nodal metastases.
elevated in patients with MTC, as with many other NETs, but calcitonin The optimal timing of prophylactic thyroidectomy depends on the
and CEA are the most useful for following MTC patients in the risk level of the RET mutation. In general, current guidelines recom-
long term.192,199,212 mend operating on children with MEN2A and familial MTC by age
Genetic testing plays an important part in the initial management 5 years, whereas those with MEN2B should be operated on before 6
because it can be used to identify familial disease and to risk-stratify months of age.225 Prophylactic surgery should consist of at least a total
patients. Germline mutations in the RET gene characterize familial thyroidectomy. The role of prophylactic lymph node dissection in
disease.213 A small percentage of apparently “sporadic” disease will familial disease remains controversial. Lymph node metastases are
also carry germline RET mutations, but truly sporadic cases frequently present in 6% of screened patients,226 and therefore some argue that
harbor somatic RET mutations. For sporadic MTCs lacking RET prophylactic central lymph node dissection should be performed.
mutations, 18% to 80% have been found to have HRAS, KRAS, or Opponents to this approach state that with normal preoperative
NRAS mutations.214–216 In sporadic MTC, presence of RET, especially ultrasound findings, normal calcitonin level (basal and/or stimulated),
RET codon M918T, mutation usually predicts a poor prognosis.190,217,218 and a normal CEA level, the risk of occult nodal disease is very low
1088 Part III: Specific Malignancies

and does not outweigh the risks of a central neck dissection such as Brazil, with a 10 to 15 times higher incidence in children and with
permanent hypoparathyroidism.9,221,226 Because any complications the majority of sporadic ACCs possessing a germline TP53 (R227H)
resulting from prophylactic surgery become lifelong problems for the mutation.238–240
patient, experienced surgeons should perform prophylactic surgery.
Before proceeding with surgery, the surgeon should screen patients Pathogenesis
with hereditary disease for associated conditions such as PCC (MEN2A
and MEN2B) and hyperparathyroidism (MEN2A).220,226 Most ACCs arise sporadically, and the pathogenesis is not completely
All patients will require thyroid hormone replacement once the understood. It is not known if ACC develops from hyperplastic or
thyroid is removed, but TSH suppression is not required. After surgery, adenomatous adrenal nodules. However, several genetic alterations
the next phase of treatment is surveillance. This begins 2 to 3 months have been identified; the most common mutation involves overexpres-
postoperatively with a new baseline calcitonin and CEA measurement. sion of the insulin-like growth factor (IGF) gene. Next-generation
If the calcitonin is undetectable, these patients can be followed with sequencing in 1051 children with cancers revealed that 27 (69%) out
yearly calcitonin measurements. Imaging is undertaken when the of 39 patients with ACCs possessed a TP53 mutation.241 In this study,
calcitonin level rises. family history was not a predictor of an underlying predisposition
A spectrum of disease severity exists for both hereditary and sporadic syndrome in most patients.241 β-Catenin CTNNB1 was associated
MTC, and therefore the natural history of MTC varies widely. Distant with poor outcome and decrease in overall and disease-free survival
metastases in the lung, liver, or bone can arise and lead to death quite in patients with ACCs.242 Another potential tumor suppressor gene,
quickly. On the other hand, many patients live with a large tumor ZNRF3, was identified in 21% of patients with ACCs with use of
burden and very high calcitonin levels with few symptoms. Others exome sequencing and single-nucleotide polymorphism (SNP) array
develop intractable diarrhea. In this case, cytoreductive surgery227 or analysis.243 Furthermore, tumors containing hypermethylated DNA
somatostatin analogues such as octreotide can palliate severe symp- have also been found to be associated with a poor prognosis.244 Other
toms.227 Overall 10-year survival rates have been reported as 75% and growth factor and growth factor receptor mutations have also been
95% for tumor confined to the thyroid and regional stage disease, identified in sporadic ACC.235,245 Familial tumor syndromes that involve
respectively, in a SEER registry study.192 In this study, age and stage ACC include MEN1 (11q13), Li-Fraumeni syndrome (17p13),
were found to be stronger predictors of survival in univariate analysis.192 Beckwith-Wiedemann syndrome (11p15.5 and 15q11–13), and Carney
Furthermore, calcitonin doubling times of less than 6 months indicate complex (17q23–24 or 2p16).235,246,247
worse prognosis, with 5-year and 10-year survival rates of 25% and
8%, respectively.228 Conventional chemotherapy regimens with Clinical Presentation
doxorubicin, dacarbazine, capecitabine, and 5-fluorouracil (5-FU) have
demonstrated limited efficacy in patients with MTC. According to Most ACC patients are asymptomatic until the tumor reaches a size
the revised ATA guidelines, systematic chemotherapy should not be that causes compression of nearby structures, local invasion, or distant
used as a first-line treatment.190 Newer, targeted therapies block the metastasis. Although symptoms are vague, they can include fever,
RET receptor tyrosine kinase or its multiple downstream pathways, early satiety, weight loss, pain, anemia, nausea, and fatigue. The presence
such as the extracellular signal-related kinase (ERK), phosphatidylino- of these symptoms in the setting of ACC is ominous. The mean
sitol 3-kinase (PI3-K)/Akt, p38 mitogen-activated protein kinase duration from the onset of symptoms to the diagnosis of ACC varies
(MAPK), and c-Jun N-terminal kinase pathways.106,199,229 Some of from 6 to 16 months and appears to be independent of whether the
these tyrosine kinase inhibitors inhibit multiple signaling pathways tumor is functional.234–236
simultaneously.106,230 These targeted therapies have been evaluated in In 40% to 60% of the patients, ACC results in increased hormone
multicenter trials.230,231 production (functional) that may cause symptoms and assist in cor-
The FDA has approved one of these targeted therapies, vandetanib, rectly diagnosing these aggressive tumors. Because most ACCs are
for treatment of metastatic MTC. Vandetanib is a small-molecule inefficient in mature steroidogenesis, they can secrete a variety of
inhibitor of the VEGF receptor, epidermal growth factor (EGF) receptor, steroid precursors that result in clinical or subclinical syndromes. As
and the RET tyrosine kinase.232 In a randomized controlled clinical many as 75% of the ACCs are associated with subclinical Cushing
trial, patients treated with vandetanib experienced a median PFS of syndrome, which can be diagnosed with biochemical testing.245,248
22.6 months compared with 16.4 months in patients treated with Clinical Cushing syndrome with the classic symptoms may be present
placebo.230 Another FDA-approved tyrosine kinase inhibitor is in up to 50% of the patients with functional ACC. Virilization caused
cabozantinib, which targets VEGFR1 and VEGFR2, c-MET, and by excessive androgen steroids or androgenic precursors is identified
RET. In a phase I/II trail of 35 patients with MTC, 17 patients had in approximately 20% of the patients, and the combination of clinical
a partial response.233 Patients on tyrosine kinase inhibitors should be Cushing syndrome and virilization is present in 10% of the functional
monitored for hypothyroidism.190 Important to note, none of these ACCs. Cushing syndrome with virilization almost always indicates
agents have been shown to prolong overall survival. More research is ACC. Other clinical manifestations include feminization (5%–8%),
currently being conducted on other potential therapies. hypoglycemia (<5%), and hypokalemic alkalosis (<5%).235,249 Men may
have gynecomastia or low libido owing to estrogen overproduction by
ACC.250 Eighty percent of children have virilization; 6% have isolated
ADRENOCORTICAL CANCER glucocorticoid excess.239,251
Incidence Metastases are present in 20% to 50% of the patients with ACC
at the time of diagnosis, with lungs (40%–50%), liver (40%), and
Adrenocortical cancer (ACC) is a rare endocrine malignancy with an lymph nodes (20%–30%) being the most common sites. Less common
incidence of 1 to 2 per million people; it causes less than 0.2% of all sites include the bones, spleen, pancreas, and diaphragm.235,249,252
cancer deaths.234,235 After ATC, ACC is the second most aggressive
endocrine cancer. Although ACC can develop at any age, it occurs in Diagnosis
a bimodal age distribution: in patients younger than 5 years, and those
in the fourth and fifth decades of life.236 In general, ACC in adults The correct diagnosis of ACC is important for appropriate management;
is more aggressive than in children, with rapid disease progression. however, preoperative diagnosis of ACC is complex. At present, the
Women are affected more than men, at a ratio of 1.5 : 1, and are diagnosis relies on clinical assessment, urinary and plasma biochemical
more likely to have a functional tumor.237 The incidence of ACC tests, and imaging studies. Clinical suspicion should be raised in any
also varies among countries. The highest incidence is in southern patient with adrenal Cushing syndrome with an adrenal mass, increased
Cancer of the Endocrine System  •  CHAPTER 68 1089

urinary 17-ketosteroids, and age younger than 20 years, or in patients


with an adrenal mass, increased urinary 17-ketosteroids, virilization Primary Disease
or feminization, weight loss, anemia, or fever.235,249 Surgery
Hormonal workup includes plasma adrenocorticotropic hormone, Surgical resection of the primary tumor is the only means to achieve
cortisol, and urinary 17-ketosteroids, with or without a dexamethasone cure, and every attempt must be made to identify the patients who
suppression test. Adrenal incidentalomas should also be evaluated are candidates for curative resection.235,248,252 Patients with known or
with plasma or urinary metanephrines to rule out PCC, and with suspected ACC should be referred to highly experienced surgeons.
renin and aldosterone levels in patients with hypertension to rule out The surgeon can evaluate the size and local invasiveness of the tumor
aldosteronoma.253 and decide on the approach and extent of surgery. Complete hormonal
The role of imaging in the evaluation of adrenal mass is valuable assessment should be obtained before surgery in order to determine
for assessing the tumor size and other specific imaging characteristics. the functional status of the adrenal lesion. Tumors that are localized
It has been demonstrated that 92% of the ACCs are larger than to the adrenal gland should be resected with adequate margins. Resection
6 cm254 and that the risk of malignancy increases with adrenal tumor can be performed via an open (anterior or posterior) or a laparoscopic
size as follows: 2% for tumors smaller than 4 cm, 6% for tumors that approach. Although the role of laparoscopy for the resection of ACC
are 4 to 6 cm, and 25% for tumors larger than 6 cm.248,255 Furthermore, is controversial, surgeons are expanding the indications for laparoscopy
adrenal masses that increase in size within a 6-month period are also for localized tumors with comparable outcomes.266 At present there
suggestive of malignancy.235 CT is the most useful imaging study to are no randomized controlled trials that evaluate the role of laparoscopic
evaluate adrenal tumor size, adjacent organ involvement, and resect- surgery for ACC.
ability. Irregular borders, heterogeneity, calcifications, radiodensity Before incision, patients should be given a stress dose of steroids
greater than 25 Hounsfield units (HU), delayed washout of less than because the contralateral gland may be suppressed.235 At surgery, every
50%, and extension to the inferior vena cava all suggest malignancy. attempt must be made to achieve clear surgical margins, and, in cases
In the largest reported series evaluating CT imaging for ACC, the of locally advanced disease, en bloc resection of involved organs should
authors found that improved diagnostic accuracy could be obtained be performed. Tumor spillage and incomplete resection are associated
by selecting a radiodensity cutoff for adrenal lesions of 13.9 HU at with a poor prognosis.267 Regional lymph node dissection should be
CT imaging. Sensitivity was 100% and specificity was 68%.256 In performed for involved lymph nodes, and such involvement should
addition, when performed correctly, intravenously administered contrast be assessed in every patient with known ACC.
material washout of less than 50% carries a sensitivity and specificity Advanced ACCs may involve the kidney, pancreas, spleen, liver,
of 100%.235,249,253,257 With gadolinium-enhanced magnetic resonance diaphragm, renal vein, and inferior vena cava. Tumors may even
imaging (MRI), malignant lesions show rapid and marked enhancement extend through the inferior vena cava to the right atrium and the
and a slower washout pattern on T2-weighted images. Other suspicious superior vena cava. For such advanced tumors, multidisciplinary teams
features at MRI include peripheral enhancement, central necrosis, are of the utmost importance, because cardiopulmonary bypass or
and internal hemorrhage. The overall MRI sensitivity and specificity hypothermic circulatory arrest is often required.234,248,249 Table 68.6
are 81% to 89% and 92% to 99%, respectively.258 outlines the staging of ACC. There is no consensus yet about routine
Two other imaging modalities show encouraging results. The lymphadenectomy for patients with ACCs. A study of 283 patients
sensitivity of FDG-PET was reported to be 97% after a systematic with completely resected ACCs found reduced risk of recurrence
review that included 21 studies for identifying ACC, with no difference and disease-related death among patients who underwent routine
reported between FDG-PET and FDG-PET/CT.259 C-metomidate-PET lymphadenectomy.268
(MTO-PET) is another emerging adrenal imaging technique with Postoperative surveillance should be continued for many years
similar accuracy.260,261 Another imaging technique is (123I)iodometomi- because recurrences have been documented as long as 12 years after
date (IMTO) scanning, which has the ability to reveal adrenal lesions resection. For patients with elevated dehydroepiandrosterone sulfate
with a sensitivity and a specificity of 89% to 85% and 38% to 100%, (DHEAS) at presentation, the postoperative levels may be used for
respectively.262,263
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.235,253,264 The Table 68.6  Adrenocortical Cancer Staging
definitive diagnosis of ACC is through pathologic evaluation. Grossly,
these are large tumors that may invade the adjacent organs and large TNM
blood vessels, such as the inferior vena cava. The tumors are yellow T T1 Size ≤5 cm, no local invasion
to tan, with areas of necrosis and hemorrhage. In the absence of T2 Size >5 cm, no local invasion
metastatic disease or local, capsular, or vascular invasion, the diagnosis T3 Presence of local invasion without involvement of
of malignancy may be difficult. The Weiss criteria incorporate nine adjacent organs
histologic features that help distinguish between malignant and benign T4 Invasion of adjacent organs
tumors. These features include high nuclear grade (III or IV), mitotic N N0 No positive lymph nodes
rate greater than 5 per 50 high-power fields (HPFs), atypical mitoses, N1 Positive lymph nodes
diffuse architecture, microscopic necrosis, 25% or fewer clear cells,
M M0 No distant metastases
capsular invasion, sinusoidal invasion, and venous invasion. A tumor
M1 Distant metastases
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 STAGING
molecular markers such as microRNAs, IGF2 overexpression, increased
I T1, N0, M0
Ki-67, and specific genetic variations may prove to be a more accurate
diagnostic tool.236,245,248,264 Ki-67 tumor expression greater than 10% II T2, N0, M0
positively correlates with a worse prognosis.265 III T3, N0, M0
T1-2, N1, M0
Treatment IV T4, N0, M0
T3, N1, M0
The management of ACC should involve a multidisciplinary approach T1–4, N0–1, M1
and is tailored to each patient’s disease status.
1090 Part III: Specific Malignancies

surveillance. Patients with nonfunctional tumors should be evaluated more tolerable than with ketoconazole, and metyrapone is also a
at regular intervals with physical examination and CT scan.234,235,249 cytochrome P450 inhibitor. Etomidate also inhibits 11β-hydroxylase
and cholesterol side-chain cleavage and thus reduces cortisol and
Mitotane aldosterone synthesis.235 The role of IGF2 in the progression of ACC
Mitotane (o,p′-DDD or 1,1-dichloro-2[o-chlorophenyl]-2-[p- and its overexpression by ACCs has been clearly defined. Therefore,
chlorophenyl]ethane) is a pesticide isomer that is directly toxic to therapeutic agents targeting IGF2 have been developed. In a phase
adrenocortical cells. Its exact mechanism is yet to be elucidated; however, II trial, linsitinib, an inhibitor of the IGF1 receptor (IGF1R) and
it leads to mitochondrial disruption and adrenocortical cell necro- the insulin receptor, did not increase overall survival in patients with
sis.235,248 An inhibitory effect of mitotane of sterol-O-acyl-transferase ACCs and therefore was not recommended as a treatment option.283
1 (SOAT1) that leads to accumulation of toxic lipids has been identified
as one of the mechanisms of action of mitotane on ACCs.269 Although Recurrent or Metastatic Disease
the first use in humans was described in 1959, there are no randomized As many as 20% to 50% of patients with ACC have metastatic disease
controlled trials that have shown benefit with mitotane treatment at initial presentation; 50% to 90% of patients will experience recurrence
over controls. Retrospective studies found that mitotane improves after surgical resection.232,248,249 Patients who have resectable recurrent
recurrence-free survival in the adjuvant setting. Recurrence rate in or metastatic disease should be referred to highly experienced centers
patients treated with mitotane was 49% to 55% as compared with for excision because limited disease burden can be resected with
70% to 90% in the control groups.270,271 The effect of mitotane alone acceptable morbidity. Patients undergoing complete resection for
on disease-free survival has yet to be established. An ongoing prospective recurrent or metastatic disease have a significantly improved median
randomized study in Europe will evaluate the efficacy of adjuvant survival of 74 months, compared with 16 months for those with
mitotane therapy in patients with low to moderate recurrence risk. incomplete resection.284 Patients with unresectable recurrent or meta-
Most patients are started at a low dose of 0.5 g twice per day and static disease should be treated with mitotane alone or combination
increased to 6 g/day over 4 to 12 weeks as tolerated; the therapeutic chemotherapy even though the effect on overall survival is not proven.235
window of mitotane is narrow, and toxicity may be evident when
doses reach 8 to 10 g/day. Side effects of mitotane are predominantly Prognosis
gastrointestinal (GI), neurologic, and cutaneous. GI side effects occur
in approximately 80% of patients and include anorexia, nausea and ACC carries a poor prognosis, with a median survival of 38 months
vomiting, and diarrhea. Neuromuscular toxicity occurs in 4% to 60% and an overall 5-year survival rate of less than 40%.234,235,249 Prognosis
of patients, and the common symptoms are lethargy and somnolence. mainly depends on the tumor stage at presentation and varies dramati-
Vertigo is observed in 15% of the patients. Leukopenia and liver cally among studies. Table 68.7 summarizes the median survival and
toxicity are less common.272,273 These side effects are often severe enough 5-year survival rates for the different stages.234,235,248 A better prognosis
to require discontinuation of treatment. Tumor regression occurs in has been observed in patients with stage II ACC who received adjuvant
approximately 25% of patients, and it is rarely apparent before 6 mitotane.285 Multiple variables have been shown to portend poorer
weeks of treatment.274 outcome, including tumor size over 12 cm, age older than 55 years,
nodal or distant metastases at time of initial presentation, adjacent
Chemotherapy organ invasion, poorly differentiated histologic features, high mitotic
The role of chemotherapy for the management of ACC remains under rate, atypical mitoses, and tumor necrosis.236,238
investigation. As many as 20% of ACC patients have unresectable
tumors at presentation, and the prognosis of such patients is dismal.
Mitotane alone or with the combination of chemotherapy remains MALIGNANT PHEOCHROMOCYTOMA
the standard treatment for unresectable or metastatic disease. A recent Incidence
randomized controlled study compared 149 patients treated with
mitotane and streptozocin (M-SZ) with 148 patients treated with PCCs are rare adrenal tumors that secrete catecholamine and are
mitotane, etoposide, doxorubicin, and cisplatin (M-EDP). Patients derived from the chromaffin cells of the adrenal medulla. Paragangliomas
in the M-EDP group had a significantly higher response rate than are similar tumors that are located at extraadrenal sites along the
those in the M-SZ group (23% versus 9%) and longer median PFS sympathetic chain (organ of Zuckerkandl, pelvis, abdomen, media-
(5 versus 2 months). There was no significant difference in overall stinum, and neck).286,287 The estimated prevalence of PCC is 1 : 2500
survival (15 versus 12 months).275 to 1 : 6500 in Western countries, with an incidence of 2 to 8 per 1
million adults. The gender distribution of PCC is nearly equal, and
Radiotherapy the peak age of occurrence is the third to fifth decades of life; however,
Radiotherapy to the tumor bed has also been used as adjuvant treatment familial PCC occurs at an earlier age.286–288
after ACC resection. A small retrospective study (n = 14 in each arm) The rate of malignancy differs between PCC and extraadrenal
identified a local recurrence reduction from 79% to 19% with adjuvant paragangliomas. Although only 3% to 13% of the PCCs are malig-
radiotherapy.276 Local control was observed in 56% to 100% of patients nant, the estimated rate for paragangliomas is 15% to 35%.287,289
treated with adjuvant radiotherapy in several studies.276–280 However, Some studies have suggested that the rate of malignancy increases
this has not translated to improved survival. At present there are no with older patient age and larger tumors. Specific mutations in the
clear recommendations for radiation therapy for ACC regarding patient
selection, indications, or dosage.281
Table 68.7  Median and 5-Year Survival
Hormonal control Adrenocortical Cancer Stages
Of patients with a heavy tumor burden, 40% to 60% have symptoms
associated with excessive hormonal secretion.252,282 Several different drugs Median Survival (months) 5-Year Survival
may alleviate these symptoms. As mentioned earlier, mitotane is toxic to Stage I 61–110 62%–92%
adrenocortical cells. Ketoconazole inhibits steroid synthesis via inhibi-
Stage II 23–98 58%–87%
tion of C17-20 desmolase. Ketoconazole toxicity includes many side
effects, and because it inhibits cytochrome P450, it may interfere with Stage III 7–17 18%–40%
doxorubicin and etoposide. Metyrapone reduces cortisol and aldosterone Stage IV <12 0%–14%
production via inhibition of cortical 11β-hydroxylation. Toxicity is
Cancer of the Endocrine System  •  CHAPTER 68 1091

succinyl dehydrogenase (SDH) gene family (SDHB) are associated highly accurate for functional PCCs. The sensitivity and specificity
with a higher rate of extraadrenal tumors, and the malignancy rate of each test vary among institutions. It has been suggested that measure-
approaches 50%.287,290 ment of plasma fractionated metanephrines is the superior test.286,298,302
Other biochemical diagnostic tests include clonidine suppression test,
Pathogenesis plasma catecholamines, CgA, neuropeptide Y, vanillylmandelic acid,
and provocative testing; however, because of variable accuracy, these
Most PCCs are sporadic tumors; however, up to 24% develop from tests have not gained wide acceptance.286,298,302 To overcome the problems
hereditary germline mutations. Ten percent of PCCs are associated with with fluorometric analysis, most laboratories now measure fractionated
MEN2A or MEN2B with a mutation in the RET gene (10q11.2), and catecholamines and fractionated metanephrines.303
these are more frequently bilateral with a low (3%) malignancy rate. Imaging studies to localize the tumor should follow biochemical
A mutation in the von Hippel-Lindau gene (3p25-26) is associated diagnostic confirmation. Approximately 90% of the tumors are PCCs
with 5% malignancy rate. PCC may be a part of neurofibromatosis within the adrenal and 95% of the tumors are within the abdomen. Both
(NF) type 1 with a mutation in NF1 gene (17q11.2). The malignancy CT and MRI are highly sensitive (97% to 100%), but the specificity is
rate in these patients is 11%. The aforementioned SDH gene family only 60% to 75%. For malignant PCC, these imaging modalities may
differs in malignancy rates. Mutation in the SDHB gene (1q36.13) identify local invasion or, occasionally, distant metastases.288,298,302,304
123
is associated with a 50% to 66% malignancy rate, and mutation in I-metaiodobenzylguanidine (123I-MIBG) is a compound resem-
SDHD gene (11q23) is associated with less than 3% malignancy.287,291,292 bling norepinephrine that is taken up by adrenergic tissue. It is highly
specific (>96%) and may be used when CT or MRI findings are
Clinical Presentation negative or to identify extraadrenal metastases when malignancy is
suspected.305 OctreoScan and FDG-PET constitute the third-line
Malignant PCCs are histologically and biochemically similar to benign imaging modalities and are of benefit when malignancy is suspected.
ones. Metastatic spread is the only true indicator of malignant behavior PET with 6-(18F)-fluorodopamine ([18F]-DOPA) can be used to detect
according to 2004 World Health Organization (WHO) criteria.293 extraadrenal PCCs and neck paragangliomas; however, this imaging
More than 50% of malignant PCCs may be nonfunctional, and their modality is not yet available at all centers.302,304
diagnosis is made only after resection and pathologic evaluation. Although the risk of malignancy increases with size for all PCCs,
Functioning malignant PCCs manifest with the same secretory pattern size alone is not a reliable predictor of malignancy in local disease.
as benign tumors. The classic triad of symptoms in patients with PCC Because pleomorphism, nuclear atypia, abundant mitotic figures, and
includes episodes of headache, tachycardia, and sweating. Most of the even capsular invasion may be observed in benign PCCs, the diagnosis
patients with PCC do not have all three classic symptoms. Paroxysmal of malignancy may be difficult at pathologic assessment. Several other
hypertension may be observed in nearly 50% of the patients.294,295 methods have been evaluated to differentiate between benign and
Other signs and symptoms include papilledema, weight loss, polyuria, malignant PCC. CgA has been shown to be significantly increased
polydipsia, orthostatic hypotension, hyperglycemia, leukocytosis, in malignant tumors. Several genetic markers such as SDHB, MIB-1,
increased erythrocyte sedimentation rate, constipation, and psychiatric nm23-H1, tissue inhibitor of metalloproteinase-4 (TIMP-4), BRMS-1,
disorders.296–298 Cardiomyopathy and pulmonary edema may be caused TXNIP, CRSP-3, and E-cadherin (E-cad) are also dysregulated in
by the catecholamine excess.299 Symptoms and signs may be exacerbated malignant PCC. Tissue markers such as cyclooxygenase-2, secretogranin
or triggered by stress or consumption of tyramine.296,298 As many as II-derived peptide, heat shock protein 90, hTERT, and Ki-67 have
50% of patients with MEN2 are asymptomatic at presentation, and all been evaluated with encouraging results.286,287,306 Thompson created
a similar finding has been observed with PCC associated with von a PCC of the adrenal gland scaled score (PASS) based on histologic
Hippel-Lindau disease, in which 35% of these patients lack symptoms. features. The following items and their values (in parentheses) were
This low rate of symptomatic patients may be a result of early detection given to each tumor. Together, if present, these features would determine
by screening of at-risk patients.300,301 the PASS: large nests or diffuse growth greater than 10% (2), central
PCC should be suspected in patients who have one or more of or confluent tumor necrosis (2), high cellularity (2), cellular monotony
the following: (2), tumor cell spindling (2), more than 10-HPF mitotic figures (2),
atypical mitotic figures (2), extension into adipose tissue (2), vascular
• Classic triad spells
invasion (1), capsular invasion (1), profound nuclear pleomorphism
• Self-limited palpitations, tremor, or pallor
(1), and nuclear hyperchromasia (1). The PASS was later validated
• Hypertension resistant to medical treatment or onset before age
in another cohort, and it was identified that all cases with a PASS of
20 years, with or without atypical diabetes mellitus
less than 4 were benign, whereas all malignant cases had a PASS
• Hypertensive response to stress (surgery, anesthesia, other procedure)
greater than 6.307,308
• Presence of other syndrome-associated disease in patient of family
member
• MEN2—MTC, primary hyperparathyroidism, mucosal neuromas; Treatment
marfanoid habitus
The management approach to malignant PCCs generally follows the
• von Hippel-Lindau—angiomatosis, hemangioblastomas, pancreatic
algorithm for ACC.
cysts, café au lait spots, renal cell carcinoma
• NF1—neurofibromas, café au lait macules, freckles, optic glioma, Surgery
Lisch nodules, osseous lesions
Surgery should be offered to all patients with PCC unless the tumor
• SDH—renal cell carcinoma, GI stromal tumors, testicular seminoma
is deemed unresectable. At surgery, every attempt must be made to
• Carney syndrome—pulmonary chondromas and gastric stromal
resect the entire adrenal gland with clear margins. Surgical debulking
tumor
is considered the mainstay of therapy even for metastatic PCCs, with
• Idiopathic dilated cardiomyopathy
the rationale of reducing the circulating catecholamine levels and to
• Incidental adrenal mass
increasing the uptake of future 131I-MIBG treatment.286,289,297,304
Preoperative treatment should include fluid hydration to increase
Diagnosis intravascular volume and α- blockade to control hypertension. This
treatment should be initiated 10 to 14 days before surgery and to
The diagnosis of PCC is confirmed with the measurement of urinary the point of mild orthostatism; β-blockade may be added in patients
or plasma fractionated metanephrines and catecholamines, which is with tachycardia.
1092 Part III: Specific Malignancies

Adrenalectomy should be performed in patients with PCC, and patients with malignant PCC were treated at the National Institutes
extraadrenal resection should be offered to patients with paragangliomas. of Health (NIH) with combined cyclophosphamide, vincristine, and
The transabdominal laparoscopic approach is the most commonly dacarbazine (CVD therapy). In a 22-year follow-up, the tumor response
used, and posterior retroperitoneal surgery may be performed for rate was 55% and the biochemical response was 72%. Patients who
smaller bilateral tumors. Highly experienced surgeons may perform are candidates for CVD therapy should be started on α-blockade,
laparoscopy, and it can be safely completed even for tumors larger because the treatment causes catecholamine release. Because CVD
than 6 cm.309 A low threshold for open surgery should be practiced therapy has no proven survival benefit, it is reserved for patients who
for locally advanced tumors and paragangliomas in complex loca- were unresponsive to 131I-MIBG or symptomatic patients with unresect-
tions.294,310 Minimally invasive robotic adrenalectomy has been used able disease.286,287,315 A systematic review and meta-analysis of four
with comparable outcomes to laparoscopy. Surgical survival rates are studies that evaluated patients treated with CVD for malignant PCCs
98% to 100%; however, the morbidity associated with surgery is more and paraganglioma suggested that a partial response on tumor volume
common, and adverse perioperative complications occur in 23% to can be achieved in 37% of patients and catecholamine suppression
37% of the patients.289,309,311,312 can be achieved in 40% of patients.316
Laparoscopic transabdominal adrenalectomy begins with placement
of the patient in a lateral decubitus position. Pneumoperitoneum is Radiotherapy
established to a pressure of 15 mm Hg, and three or four trocars are Malignant PCC is considered a radioresistant tumor; however, the
introduced into the subcostal space, from the midclavicular line to main benefit of radiation therapy is to control pain and symptoms
the posterior axillary line. associated with bone and lymph node metastases. CyberKnife radiation
For tumors located on the right side, the liver is first mobilized to and radiofrequency ablation are the most commonly used methods
facilitate exposure of the adrenal gland and the adrenal vein. The for bone metastases, but more studies are needed to determine their
dissection of the posterior peritoneum begins superiorly and along efficacy in malignant PCC.287,317
the medial border of the adrenal gland. The inferior vena cava is Radiolabeled somatostatin analogue (DOTA-Tyr[3])-octreotide
identified and the lateral border is dissected to identify the short right (DOTATOC) has shown some response with no major adverse events.
adrenal vein entering the inferior vena cava. Once the adrenal vein It may be used in patients with somatostatin receptor–positive PCC.318
has been identified, the anesthesiologist should be notified before it
is divided because this may result in hypotension. The remaining Hormonal Control
retroperitoneal attachments are dissected with ultrasonic shears. The Patients with unresectable, recurrent, and metastatic disease may have
adrenal gland with the tumor is then removed via an endoscopic bag severe effects from the excessive catecholamine secretion. These patients
through the 10-mm port, and the tumor is examined on the back have several treatment options. Phenoxybenzamine is an irreversible,
table to ensure complete excision. long-acting, nonselective, α-adrenergic receptor blocker that is mostly
Laparoscopic left adrenalectomy follows the same access approach, used for preoperative preparation of these patients, but it can also be
but on the left side. On this side, the spleen is mobilized so that the used for symptom control. Nicardipine is a long-acting calcium channel
spleen and the distal pancreas fall medially to facilitate exposure of blocker that blocks the transport of norepinephrine-mediated calcium
the adrenal gland. Sometimes the splenic flexure of the colon also into vascular smooth muscle. Metyrosine inhibits the synthesis of
requires mobilization to expose the adrenal gland. A relatively avascular catecholamines by blocking the enzyme tyrosine hydroxylase. The use
plane between the pancreas and adrenal gland is dissected, and the of one of these drugs, or a combination, may contribute to symptom
borders of the adrenal gland are identified. The left renal vein drains control in patients with functional, unresectable, malignant PCC.289,304,319
into the renal vein. This vein is carefully dissected and divided. The
remainder of the gland is dissected free from the surface of the kidney Future Drugs
with ultrasonic shears as on the right side. The discovery of molecular pathways and genetic mutations that
Resectable recurrent disease or isolated metastases are optimally characterize malignant PCC has resulted in several phase I and phase
treated with surgical resection or debulking of tumor burden, especially II clinical trials. Multikinase inhibitors, tyrosine kinase receptor
if the patient is symptomatic.286,304 inhibitors, mammalian target of rapamycin (mTOR) inhibitors,
131 hypoxia-inducible factor (HIF) inhibitors, antiangiogenic agents, and
I-MIBG HER-2/neu inhibitors are among the agents that are currently under
MIBG is structurally similar to noradrenaline and is thus concentrated investigation. Treatment of a series of 17 patients with metastatic
into chromaffin cells. MIBG may be used for imaging, and the addition PCCs and paraganglioma with sunitinib, a tyrosine kinase inhibitor,
of RAI–labeled MIBG can be an effective therapeutic option. This monotherapy resulted in a partial response of 21% and disease stabiliza-
mode of therapy is completely useless in malignant PCCs that do not tion of 36%.320 An ongoing open-label trial of sunitinib in patients
take up MIBG. Important to note, external beam radiation abolishes with advanced malignant PCC or paraganglioma (SNIPP trial) should
the ability of malignant PCC to take up MIBG, and therefore patients provide further information about the role of sunitinib in the treatment
who have been treated with external beam radiation cannot be treated this aggressive disease. Clearly, novel treatments are needed for the
with MIBG. Tumor response after 131I-MIBG treatment is 30% to treatment of patients with malignant PCC and paraganglioma.287
67% and biochemical response is 45% to 67%. Symptomatic relief
may be achieved in 76% to 89% of treated patients. 131I-MIBG Prognosis
treatment can be repeated at 6-month intervals.313 The effect of
131
I-MIBG on survival is not clearly established because it is clearly Because metastases or local recurrences after resection of “benign”
not curative. Its high side effect profile, mainly myeloablative effects, disease may occur even 20 years after resection, long-term follow-up
makes it less than ideal, but it still remains the first line of adjuvant is indicated in all patients with PCC. Plasma metanephrine measure-
therapy.286,287,289 However, treatment with 131I-MIBG in patients with ments should be obtained at regular intervals for life. In high-risk
malignant PCC that take up MIBG has been shown to be effective patients, these intervals should initially be shorter.286
in alleviating symptoms by suppressing catecholamine secretion in It is difficult to predict the prognosis of patients with malignant
metastatic tumors.313,314 PCC. Five-year survival rates are only 20% to 50%. Ten-year survival
rates of 25% have been reported.321 A worse prognosis has been observed
Chemotherapy in patients with malignant PCC as compared with malignant para-
Systemic chemotherapy should be reserved for patients with unresect- ganglioma patients.322 Survival rates decrease with increasing tumor
able, metastatic, or rapidly progressing malignant PCC. In 1988, 18 size; however, there is no difference in overall survival between malignant
Cancer of the Endocrine System  •  CHAPTER 68 1093

PCC and paragangliomas. Prognosis is also affected by tumor size, is defined as one MEN1 carrier plus at least one first-degree relative
metastatic tumor burden, rate of progression, and location of with at least one of the three main endocrine manifestations.
metastasis.286,288,307,323,324 Genetic testing is recommended for individuals who meet clinical
criteria for familial MEN1 or if sporadic MEN1 is suspected because
MULTIPLE ENDOCRINE NEOPLASIA of early-onset hyperparathyroidism or the constellation of endocrine
SYNDROMES tumors. Relatives of known MEN1 carriers should also be tested.
Because the sensitivity of DNA sequencing tests is approximately
Clinical recognition of MEN syndromes first began in the 1900s. In 70% to 90%, negative genetic testing cannot exclude the syndrome.
the past 10 to 15 years, the molecular underpinnings of each subtype Furthermore, at least 10% of MEN1 cases arise de novo.334 In the
have been identified, allowing precise genotype-phenotype associations absence of positive genetic testing results, MEN1 can be diagnosed in
and genetic testing. Prophylactic surgery plays a key role in the individuals with two of the three classic endocrine organs (parathyroid,
management of these syndromes, especially for MTC associated with pancreas-duodenum, or pituitary). Screening consists of biochemical
MEN2A and MEN2B. Therefore, multidisciplinary management, laboratory tests and imaging directed at the most common tumor sites.
including genetics professionals, surgeons, and endocrinologists, can It begins at 5 years of age with serum insulin, glucose, prolactin, and
optimize the management of patients with MEN syndromes and their IGF1. Brain MRI should also be performed at age 5 years and then
families. This section summarizes the key clinical features, genetics, every 3 years thereafter. Annual calcium and parathyroid hormone
and management issues for the three MEN syndromes: MEN1, (PTH) levels are added to the battery of laboratory tests beginning at
MEN2A, and MEN2B. More detailed discussion of the medical and age 10 years.335
surgical management can be found in each of the organ-specific
sections. Treatment
Hyperparathyroidism is often the first and most common endocri-
Multiple Endocrine Neoplasia Type 1 nopathy detected in MEN1. The disease is aggressive and recurs.
Therefore, treatment consists of subtotal parathyroidectomy, with care
Wermer first reported a familial aggregation of pituitary tumors, taken to identify each gland and to leave part of the most normal-
parathyroid hyperplasia, islet cell adenomas, and peptic ulcer disease looking gland as a remnant. Cervical thymectomy is added to the
in the 1950s.325 Separately, Zollinger and Ellison reported an association procedure to clear parathyroid cell clusters that may grow and cause
between unusual small bowel ulcerations and high gastric acid secretion recurrence with time.332
along with non–insulin-secreting islet cell adenomas.326 Later, this Pancreatic and duodenal NETs such as gastrinoma arise as multiple
constellation of tumors became recognized as the MEN1 syndrome. microadenomas; therefore, historical approaches of distal pancreatec-
This syndrome is rare, occurring in 1 in 25,000 individuals.327,328 tomy or resection of palpable tumors have failed to cure gastrin
secretion.336,337 Patients should be offered surgery when biochemical
Clinical Features markers and/or imaging findings reveal tumors of appreciable size
MEN1 usually becomes clinically apparent in the 30s and 40s with (≥1 cm), because of the higher metastatic rate in larger tumors. The
hypercalcemia from primary hyperparathyroidism. Because sporadic operative procedure consists of distal pancreatectomy and enucleation
primary hyperparathyroidism typically arises in the 50s and 60s, primary of tumors in the pancreatic head. For patients with functional tumors
hyperparathyroidism in a younger patient should prompt the clinician such as gastrinomas, duodenotomy and mucosal resection of duodenal
to suspect MEN1. The penetrance of primary hyperparathyroidism tumors are added to the pancreatic portion of the procedure. In
in patients with MEN1 is 80% to 100% by age 40 years. The incidence addition, some reports have suggested open duodenotomy, often
rate of MEN1 is 1% to 18% among patients with primary hyper- preceded by endoscopic or intraoperative ultrasound examination,
parathyroidism.329,330 Tumors of the anterior pituitary occur in 10% and subtotal pancreatectomy.338–340 Pancreatic insulinomas can be
to 60% of patients, with prolactinomas being the most common. The managed with enucleation of the pancreatic head tumors plus distal
other common components of the syndrome are pancreatic islet cell subtotal pancreatectomy.341
tumors and GI NETs. Often, these are functional islet cell tumors,
such as gastrinomas (50% of individuals) or insulinomas (10% to Multiple Endocrine Neoplasia Type 2
13% of MEN1 patients). VIP–secreting tumors (VIPomas), gluca-
gonomas, and somatostatinomas have also been reported.330 Similar The first descriptions of MEN2A appeared in the 1960s and involved
to patients with sporadic gastrinoma, the duodenum is a common MTC and PCCs.342 At that time, another syndrome that included
site of gastrinoma in MEN1 patients; however, these tumors tend to mucosal neuromas was also being reported.343 This latter syndrome
be multifocal.331 Gastric carcinoids affect 10% of patients with MEN1, later became differentiated from MEN2A and classified as its own
and bronchial or thymic carcinoids each occur in 2% of carriers.330,332 syndrome, MEN2B. Both MEN2 syndromes have a combined
Thymic carcinoids are nonfunctional and tend to be aggressive. prevalence of 2.5 per 100,000.334
Although adrenal hyperplasia and benign adrenal adenomas affect
20% to 40% of carriers, malignant adrenocortical carcinomas are Clinical Features
quite rare. Both benign and malignant adrenal tumors are usually MEN2A was initially called Sipple syndrome. It is characterized by
nonfunctional in MEN1. Finally, nonendocrine tumors, such as MTC, affecting 90% to 100%190 of carriers, PCC in 50%, and
meningioma, ependymoma, leiomyoma, lipoma, and facial angiofi- parathyroid hyperplasia in 20% to 30%. Unlike in MEN1, MEN2A
bromas or collagenomas, can also be found in patients with MEN1.332 carriers have a milder form of primary hyperparathyroidism. MEN2B
The finding of more than three angiofibromas in combination with is characterized by early-onset, aggressive MTC, and PCC without
any collagenomas in a single patient is diagnostic of MEN1 with a parathyroid hyperplasia. MEN2B patients show mucosal neuromas
sensitivity of 75% and specificity of 95%.333 of the lips and tongue with a marfanoid body habitus, giving these
patients a very characteristic appearance. These patients also develop
Genetics and Diagnosis intestinal ganglioneuromas that can cause constipation and GI
MEN1 is inherited in an autosomal dominant fashion with equal sex dysmotility.332,344
distribution. Linkage studies map the mutations to the MEN1 tumor MTC is usually the first manifestation in most patients with MEN2
suppressor gene on chromosome 11q13, encoding the MENIN gene. because of its early age of onset and high penetrance. This cancer
Several inactivating mutations account for a wide variety of disease arises from diffuse C-cell hyperplasia concentrated in the upper third
patterns. Disease expression varies even within families. Familial MEN1 of the thyroid gland, and the tumors are usually multicentric. MEN2B
1094 Part III: Specific Malignancies

carriers have a more aggressive and earlier onset of MTC compared


with MEN2A carriers.335 Table 68.8  Multiple Endocrine Neoplasia
Approximately 50% of patients with MEN2A and MEN2B develop Syndromes
PCC, and the frequency depends on the specific underlying RET
Syndrome Gene Inheritance Common Manifestations
mutation.190 Although bilateral disease is common in approximately
30% to 100%, extraadrenal paragangliomas and metastatic disease MEN1 MENIN Autosomal Parathyroid hyperplasia
are rare.301,345–347 PCCs with MTC occur earlier than sporadic forms, dominant Neuroendocrine tumors of
with a mean age of presentation between 25 and 32 years.346,348 pancreas or foregut
Anterior pituitary tumors
Genetics and Diagnosis MEN2A RET Autosomal MTC
In 1987, genetic linkage studies identified the RET proto-oncogene dominant Parathyroid hyperplasia
on chromosome 10q11.2. The MEN2 syndromes result from missense Pheochromocytoma
germline mutations in this gene.349 Specific codon mutations correlate MEN2B RET Autosomal MTC
with MEN2A and MEN2B (in addition to FMTC). For example, dominant Pheochromocytoma
codon 634 mutations occur in 40% of patients with MEN2A, whereas Ganglioneuromatosis
codon 918 mutations occur in 95% of patients with MEN2B. MEN2B Marfanoid body habitus
mutations correlate with a more aggressive MTC phenotype and MTC
metastases before 12 months of age. An international consensus panel MTC, Medullary thyroid cancer.
developed a three-tier risk stratification based on specific codon
mutations. This risk stratification guides the timing of prophylactic
thyroidectomy.334,350,351 It should be noted that somatic RET mutations indicated, the patient will require lifelong mineralocorticoid and
are found in approximately 25% of patients with both sporadic MTC glucocorticoid replacement. Because these patients then develop the
and sporadic PCC.351 RET gene testing should be offered to all members complications of long-term steroid medication use and are at risk for
of MEN2 kindreds. Patients with PCC should also undergo RET addisonian crisis, some centers prefer to perform cortical-sparing
gene testing. However, diagnosis of MTC can be made in the absence adrenalectomy. Intraoperative ultrasound guides the extent of resection.
of a RET mutation in patients who have the classic clinical presentation Adrenalectomy can be performed with an open, laparoscopic, or
of MEN2.352 Furthermore, diagnosis should be suspected in patients retroperitoneoscopic approach.358,359
with a family history of MTC, younger age at presentation (<35 Table 68.8 summarizes the MEN syndromes.
years), or multicentric tumors.
Ideally, genetic screening and prophylactic thyroidectomy should CARCINOID TUMORS
preclude the need for diagnosing MTC in MEN2 kindreds. However,
FNA and biochemical testing are sometimes necessary (see section Carcinoid is an English translation of a term first used by German
on MTC). Annual biochemical screening (plasma or 24-hour pathologists in the early 20th century—karzinoide—to describe a
urinary metanephrines) and CT scans can exclude PCC in patients carcinoma-like tumor that appeared to have more indolent behavior
with MEN2.334,335 than a carcinoma.360 The term is somewhat of an anachronism now
but remains in common use for describing a well-differentiated NET
Management that secretes serotonin, which is how the term carcinoid is used in
The timing and extent of prophylactic surgery for MTC among RET this section. High-grade or poorly differentiated NETs are now classified
mutation carriers depend on the specific codon mutation. As mentioned, as neuroendocrine carcinomas. Carcinoid tumors are an important
the MTC associated with MEN2B manifests earlier and behaves more subset of a broader class of neoplasms collectively called neuroendocrine
aggressively. Therefore, patients with MEN2B should undergo pro- tumors. Other NETs include medullary carcinoma of the thyroid,
phylactic thyroidectomy before 1 year of age. If thyroidectomy occurs PCC, and pancreatic endocrine tumors. This section approaches the
after 1 year, then prophylactic central neck dissection (level VI) is carcinoid subset of NETs separately from pNETs, which confers
added to the procedure. In MEN2A, prophylactic thyroidectomy is important diagnostic, symptomatic, and therapeutic differences.
offered before age 5 years (i.e., codon 634 mutations). Surgery can
be offered earlier for higher-risk mutations. When MTC is discovered Incidence
clinically, central compartment (level VI) neck dissection is added to
total thyroidectomy. Similarly, compartment-oriented dissection is Data regarding the incidence of carcinoid tumors continue to evolve.
indicated when lymph node metastases are discovered clinically. This According to data from the SEER database, the incidence of all NETs,
may include the lateral neck compartments, if positive nodes are including carcinoid NET, pNET, and poorly differentiated NET has
discovered in these locations.332,352 The mortality from MTC was risen from 1.09 per 100,000 to 5.25 per 100,000 population. This rise
reported to be 50% of patients with MEN2B and 9.7% of patients may very well be directly attributable to increased detection because
with MEN2A in one large follow-up study.353 The 10-year survival of modern CT imaging, but the rise in NET diagnosis outstrips the
rate of MTC has been reported to be 61% to 76%, with old age at expectation even when increased detection is taken into account.361
diagnosis, extent of primary tumor, nodal disease, and distant metastasis Carcinoid tumors typically have been classified as originating in
as important prognostic factors that predict adverse outcome.352,354–356 the foregut (lung and upper GI tract and, less often, pancreas), the
PCC most commonly occurs with mutations in codons 634 and jejunum and midgut (which includes the ileum and appendix), and
918 but can occur in all MEN2-associated mutations except for codon the hindgut (colon and rectum). In a series of more than 13,000
768. PCC in patients with MEN2 is treated with adrenalectomy when carcinoid tumors reported through End Results Group (ERG), the
the disease becomes clinically evident with elevated catecholamines— Third National Cancer Study (TNCS), and SEER, 66.9% of carcinoids
that is, prophylactic adrenalectomy is not indicated. PCCs are rarely came from the GI tract, and 25.3% came from the bronchopulmonary
(~15%) the initial manifestation in MEN2 syndromes but do occur tree. Within the GI tract, 41.8% of tumors came from the small
concomitantly with MTC in 25% of patients.357 Consequently, intestine, 27.4% from the rectum, and 8.7% from the stomach. There
biochemical testing should be performed before thyroidectomy. is a slight female predominance (55.1%). White patients tended to
Although PCCs are often bilateral, they develop asynchronously. have more bronchial carcinoids than other ethnic groups, and rectal
A contralateral PCC occurs within 5 years of adrenalectomy in carcinoids seem to have been overrepresented among patients of African
approximately 30% of patients.358 When bilateral adrenalectomy is descent.362
Cancer of the Endocrine System  •  CHAPTER 68 1095

Table 68.9  Grading Proposal for NET Table 68.11  ENETS Proposal for TNM Staging of
(All Sites)236–238 NET in Small Intestine237
Grade Mitotic Count/10 HPF Ki-67 Index (%) T—PRIMARY TUMOR
G1 <2 ≤2 Tx Primary tumor cannot be assessed
G2 2–20 3–20 T0 No evidence of primary tumor
G3 >20 >20 T1 Tumor invades lamina propria or submucosa and <1 cm
T2 Tumor invades muscularis propria or >1 cm
HPF, High-power field; NET, neuroendocrine tumor.
T3 Tumor invades pancreas or retroperitoneum
T4 Tumor invades peritoneum or other organs
N—REGIONAL LYMPH NODES
Table 68.10  Staging Proposal for NET Nx Regional lymph nodes cannot be assessed
(All Sites)236–238 N0 No lymph node metastasis
Disease Stage T N M N1 Regional lymph node metastasis

I T1 N0 M0 M—DISTANT METASTASES
IIa T2 N0 M0 Mx Distant metastasis cannot be assessed
IIb T3 N0 M0 M0 No distant metastasis
IIIa T4 N0 M0 M1 Distant metastasis
IIIb Any T N1 M0
ENETS, European Neuroendocrine Tumor Society; NET, neuroendocrine tumor.
IV Any T Any N M1

NET, Neuroendocrine tumor.

Table 68.12  ENETS Proposal for TNM Staging of


Pancreatic NET236,237
Clinical Pathology and Staging T—PRIMARY TUMOR
Carcinoid tumors usually are readily identified microscopically. Tx Primary tumor cannot be assessed
Immunohistochemical staining is performed to identify and classify T0 No evidence of primary tumor
these tumors. In addition to immunohistochemical staining to reveal
T1 Tumor limited to the pancreas and <2 cm
specific tumor produced peptides, other semispecific markers, including
neuron-specific enolase (NSE)363 and CgA,364 aid in diagnosis. T2 Tumor limited to the pancreas and 2–4 cm
Appropriate TNM (tumor-nodes-metastases) assessment and discern- T3 Tumor limited to the pancreas and >4 cm or invading
ment of tumor grade are the essential components of a pathologic duodenum, bile duct
diagnosis. As mentioned earlier, nomenclature for NET as a whole T4 Tumor invading adjacent organs
has become challenging. Older nomenclature using “benign” versus
“malignant” or “typical” versus “atypical” carcinoid is thought to be N—REGIONAL LYMPH NODES
inaccurate, because all carcinoids may have some malignant potential. Nx Regional lymph nodes cannot be assessed
The WHO classification divided NET into well-differentiated endocrine N0 No lymph node metastasis
tumors, well-differentiated endocrine carcinomas (WDECs), and poorly N1 Regional lymph node metastasis
differentiated endocrine carcinomas (PDECs). In spite of this improve-
ment over older descriptions, the WHO classification still fell short M—DISTANT METASTASES
in terms of how to predict the malignant behavior of low-grade Mx Distant metastasis cannot be assessed
tumors.365 In an attempt to combine anatomic staging with tumor M0 No distant metastasis
grade, the European Neuroendocrine Tumor Society (ENETS)
M1 Distant metastasis
developed a staging and classification system for NETs that has proven
useful for prognostication at each anatomic site366,367 (Tables 68.9 ENETS, European Neuroendocrine Tumor Society; NET, neuroendocrine tumor.
through 68.12). SEER data suggest that across all stages and sites,
the 5-year survival in patients with carcinoid tumors is 67.2%. Carcinoid
tumors of the rectum typically have lower risk of recurrence and better
5-year survival (88.3%) than other GI tract carcinoids (67.5%) and the GI tract accounting for more than two-thirds of primary tumors.
bronchopulmonary carcinoids (73.5%).362 There are reports of carcinoid tumors of the bladder, breast, ovary,
Tumors of mixed histology, such as goblet cell or adenocarcinoid testis, and thymus.362 As noted earlier, biopsy confirmation, with
tumors of the GI tract, have both mucinous and neuroendocrine appropriate staging and grading, is critical because the biologic behavior
differentiation. These tumors are often more aggressive than typical of low-grade NETs is typically more indolent than that of adenocar-
GI carcinoids and require staging and management similar to those cinoma of any of the aforementioned sites.
of adenocarcinoma.368
Diagnosis
Anatomy
The diagnosis of carcinoid tumor is made by finding tumor or symptoms
Carcinoids are thought to arise from the Kulchitsky cells in the crypts related to mass effect, biologically active peptides, or urinary tumor
of Lieberkühn of the gut, and are disseminated in the endobronchial markers (Fig. 68.7). Although measurement of 5-hydroxyindoleacetic
mucosa.369 These tumors can arise from nearly any anatomic site, with acid (5-HIAA) is the most common and reproducible test for the
1096 Part III: Specific Malignancies

Symptom/finding

Diarrhea Tumor with no syndrome


Flushing
Hypotension
Rarely
Tumor location unclear Tumor location apparent
Heart disease
Pellagra
Asthma Staging
Osteoarthropathy
Contrast radiography or CT
scan of abdomen and chest
Consider: carcinoid

No elevation of urinary Urinary 24-hour Metastatic tumor Localized tumor


24-hour 5-HIAA 5-HIAA > 6 mg/24 hr
(> 30 mg/24 hr in
patients with diarrhea Tissue diagnosis Tissue diagnosis
Consider or malabsorption)
Islet tumor or carcinoid
with no serotonin Surgical resection
production (rare) Carcinoid tumor
Cure

No carcinoid syndrome Carcinoid syndrome present

Pharmacologic therapy
Tumor bulk Symptomatic liver to decrease symptoms
Observation Somatostatin analogue
symptoms metastases
?Alpha-interferon

Cytoreductive Cytoreductive surgery or Progression


surgery hepatic artery ligation or
hepatic arterial embolization
± chemotherapy
Progression Surgical therapy Nonsurgical
therapy

Metastases any site Liver mets

Cytoreductive Cytoreductive surgery or


surgery hepatic artery ligation or
hepatic arterial embolization
± chemotherapy

Progression Progression Progression

Systemic therapy, chemotherapy or biologic response


modifiers

Figure 68.7  •  Algorithm of diagnosis and treatment of carcinoid tumors. CT, Computed tomography.
Cancer of the Endocrine System  •  CHAPTER 68 1097

presence of carcinoid syndrome; significantly elevated levels of 5-HIAA Anterior Posterior


usually occur only after there is a 20% to 25% liver tumor burden.
In most laboratories, the upper limit of normal for 24-hour urinary
5-HIAA excretion is 6 to 10 mg. In one study, 5-HIAA measurements
were 100% specific and 73% sensitive for the presence of carcinoid
syndrome.370 Not all patients with carcinoid tumor have the associated
syndrome, and the sensitivity of assays for detecting the presence of
tumor alone is inadequate. Although markedly elevated 5-HIAA in
the urine is specific for carcinoid tumor, a low-level false-positive
increase of 5-HIAA may be seen in patients with noncarcinoid tumor
and after intake of certain foods (e.g., bananas, walnuts, and pecans)
and serotonin reuptake inhibitors (e.g., sertraline [Zoloft], fluoxetine
[Prozac]) and other medications (e.g., acetaminophen, salicylate,
guaifenesin).371–373 5-HIAA also may be elevated to low abnormal
levels (<30 mg) in patients with diarrhea or malabsorption from any
cause. In addition to excellent specificity and high sensitivity, 5-HIAA
measurement has a high level of consistency, both in individual patients
and among groups. In the Mayo Clinic series, the level of 5-HIAA
excretion remained constant in a group of 85 patients in whom paired
determinations were performed during a 10-day period.374 Moreover,
in any given patient, the level of 5-HIAA secretion is a relatively
accurate indicator of tumor bulk. A novel test of plasma 5-HIAA has
been developed in which results were significantly correlated with the
urine 5-HIAA.375 In a study that included 500 specimens of consecutive Figure 68.8  •  Indium-111 (111In) octreotide scan. Patient with metastatic
raised plasma CgA, urine 5-HIAA, and plasma neurokinin A (NKA), carcinoid with octreotide scan showing bony and visceral metastases with
CgA remained the best general circulating marker for NETs.376 Serum anterior and posterior views.
NSE and CgA levels have been shown to correlate with the presence
and natural history of GI NETs.363,364 For midgut carcinoids, which
produce high levels of serotonin, the sensitivity of CgA levels is only
slightly better than that of urinary 5-HIAA levels. However, for flushing.387,388 Serotonin may be responsible for intestinal hypermotility
foregut- and hindgut-derived tumors, CgA levels are highly sensitive and hypersecretion, but it probably does not cause the characteristic
compared with urinary 5-HIAA levels.377 However, sensitivity and flushing that occurs with the carcinoid syndrome.389 Vasodilation,
specificity of CgA depends on the cutoff value.378,379 Like 5-HIAA, which causes flushing, can be caused by one or more substances released
CgA may be elevated in nontumoral conditions such as atrophic by the tumor cells, including bradykinin, substance P (midgut carci-
gastritis, proton pump inhibitor (PPI) use, liver or kidney dysfunction, noids), tachykinins, and prostaglandins. The symptoms of the carcinoid
and inflammatory bowel disease.377 CgA may be an important early syndrome vary in frequency. Flushing is most frequent, followed by
biomarker of response to some types of therapy. diarrhea, heart disease, and bronchoconstriction. Presence of carcinoid
As CT and MRI scanning technologies improve, the ability to syndrome and heart disease are adverse prognostic indicators for
localize even small tumors has improved considerably and may allow survival.390
for more precise imaging of primary and metastatic lesions.380 Most
carcinoid tumors are highly vascularized and enhance with iodinated Flushing
contrast agent during the early arterial phase.381 However, for metastatic Two types of flushing are accepted as accompanying the usual metastatic
liver disease, MRI is probably the most sensitive method.382 Localization ileal carcinoid. It is thought that any of the activities that drive plasma
of carcinoid and islet cell tumors has been investigated by nuclear catecholamines, which instigate the flushing, may be mediated, in
medicine techniques (Fig. 68.8).383,384 The sensitivity of somatostatin part, by peptides such as the tachykinin substance P or the amine
receptor or octreotide scanning (OctreoScan) is reported to be up to serotonin. Stressors associated with hypercatecholemia include exercise,
87%, with a positive predictive value of 100%, making it a useful excitement, emotion, ethanol, and decongestants such as ephedrine. One
imaging study for carcinoid tumors.383 Early indication of a possible type of flushing is red and diffuse, involving the face and upper body;
response to octreotide-based therapy (see later) may also increase the it is of short duration and can be provoked by alcohol, excitement,
usefulness of octreotide scanning. Recently developed and approved emotional stress, and catecholamine release.391,392 The other is more
by the FDA, gallium-68 (68Ga)–DOTATATE PET-CT imaging has prolonged, produces venous dilation and a purplish hue about the
a higher spatial resolution than conventional OctreoScan and better face, and can give rise to permanent dilation of facial veins and even
sensitivity in detection of small tumors.385 telangiectasia. This flush is more commonly precipitated by alcohol
ingestion. Because infusion of serotonin does not cause either type of
Symptoms flush, it has been suggested that the kinins cause this symptom.393 Brief
flushes may be caused by catecholamine-induced release of vasoactive
Carcinoid Syndrome substances such as kallikreins; these flushes can be blocked effectively
Many patients with metastatic carcinoid tumor manifest the signs by α-adrenergic blocking agents.394 Carcinoid of the foregut produces
and symptoms of abnormal hormone production—the malignant a more intense and erythematous flush (as seen in a serotonin burst),
carcinoid syndrome.386 Serotonin (5-hydroxytryptamine [5-HT]), sometimes associated with itching, conjunctival suffusion, and facial
synthesized by the tumor from tryptophan and metabolized to 5-HIAA, edema suggestive of histamine release. Occasionally, gastric carcinoids
which appears in the urine, is particularly important because urinary cause an urticarial reaction, which may be inhibited by the histamine
5-HIAA levels are used to monitor the course of carcinoid syndrome. H1- and H2-receptor antagonists diphenhydramine and ranitidine.395
However, the relation of serotonin levels to symptoms of the clinical
carcinoid syndrome is uncertain. Carcinoid tumors also release the Diarrhea
enzyme kallikrein, which acts on α2-globulin to produce bradykinin The diarrhea of carcinoid syndrome does not necessarily correlate
and its precursor, lysyl-bradykinin, both of which can induce with flushing. Diarrhea appears to be related to increased gut motility
1098 Part III: Specific Malignancies

rather than to secretion of fluids. Diarrhea of the carcinoid syndrome such complications and an aggressive approach in considering surgical
rarely is of high volume and therefore typically requires only mild palliation. Disease-specific 10-year survival rates of small intestine
palliative antidiarrheal therapy. Although abdominal cramping can NETs were reported in a SEER cancer registry study, ranging from
be associated with this form of diarrhea, other possibilities for abdominal 95% for stage I to 42% for stage IV.407 However, even with metastatic
pain must be considered, including intermittent partial small bowel disease, 5-year and 10-year survival rates of 40% to 85% and 40%
obstruction secondary to mesenteric fibrosis or bowel obstruction to 60%, respectively, have been reported.407–411
secondary to tumor bulk. Management of hepatic metastases is critical. The goal of therapy
is to control symptoms and improve quality of life. In many patients,
Heart Disease bulky hepatic metastases constitute most of the tumor burden, so
The cardiac disease associated with the carcinoid syndrome is an tumor reduction may, at the least, diminish production of peptides
endomyocardial fibrosis typically involving the right side of the heart, that promote the carcinoid syndrome, in addition to extending sur-
although left-sided lesions have been described in the setting of patent vival.412 Historically, resection is recommended if at least 90% of the
foramen ovale.396 Fibrotic deformation of the tricuspid and pulmonary tumor can be feasibly removed. The indications to proceed with hepatic
valves usually leads to pulmonary stenosis and tricuspid insuffi- metastasectomy are more liberal in carcinoid tumors than in metastases
ciency.397,398 Carcinoid heart disease is a late complication of carcinoid from other solid tumors. In a very large multiinstitutional series of
tumors, with the disease developing after an average of 5 years from 339 patients who underwent hepatic resection for NET liver metastasis,
diagnosis of carcinoid tumor. The prevalence of carcinoid heart disease 5-year and 10-year survival rates were 74% and 51% respectively.413
has fallen from 50% to 70% of patients with carcinoid syndrome to This is consistent with the findings of a meta-analysis of cytoreductive
20%, likely as a consequence of the use of octreotide and other agents.398 partial hepatectomy for carcinoid metastases, in which surgical debulk-
Screening patients with carcinoid tumors with brain natriuretic peptide ing yielded a 5-year survival rate greater than 70% and resolution of
or echocardiogram can be considered if symptoms of heart disease carcinoid symptoms in 86% of patients.414 In patients whose extrahepatic
are present.399–401 disease has been eliminated, liver transplantation may be an option
Other, relatively rare signs and symptoms may be associated with as well.414a,414b
carcinoid syndrome. Bronchoconstriction may occur in both pulmonary
and extrapulmonary carcinoid, and usually is associated with flushing. Radiation Therapy
The classic triad of dermatitis, dementia, and diarrhea seen with pellagra Radiation therapy seldom is used to treat carcinoid tumor or carcinoid
occasionally has been identified. This syndrome is secondary to niacin syndrome. Patients with carcinoid syndrome often have extensive
deficiency as a result of shunting of dietary tryptophan from niacin hepatic metastases, and the dose-limiting toxicity of hepatic radiation
synthesis to indole synthesis. It is very rare in the United States and limits its usefulness. Trials of stereotactic body radiotherapy (SBRT)
other developed countries because of better overall nutrition compared have shown that doses up to 60 Gy can be given in a focused way to
with developing and underdeveloped countries. It is treated simply oligometastatic disease.415 Trials looking at disease-specific survival
with nicotinamide from nonprescription vitamin supplements. after SBRT are ongoing.
Palliative treatment of bone metastases is an indication for radiation
Therapy therapy.

Management of carcinoid tumors depends not only on traditional Antihormonal Therapy


methods of dealing with the tumor itself, but also on management When the symptoms attributed to serotonin are mild, they can be
of associated medical problems typically caused by overproduction of managed successfully over lengthy periods with simple measures, such
hormonally active peptides. When curative surgery is not possible, as administration of opiates and diphenoxylate hydrochloride with
surgical debulking may result in symptomatic improvement. atropine.
About 80% of carcinoid tumors express somatostatin receptors,
Surgery which can be determined with OctreoScan or 68Ga-DOTATATE
Surgery has an important role throughout the course of carcinoid tumor PET-CT. Intravenous or subcutaneous somatostatin inhibits carcinoid
management. Resection of the primary tumor and of associated resect- flush and significantly improves diarrhea in over 80% of patients416
able nodal metastases is primary therapy.402 Surgical approaches clearly but is not practical for therapy because it has a half-life of less than
differ by site. Carcinoids of the appendix commonly are discovered 2 minutes.417 Synthetic octapeptide analogues of somatostatin, octreotide
incidentally. Most are smaller than 1.0 cm and are cured by surgical acetate, lanreotide, and vapreotide have longer half-lives and can be
resection. In the Mayo Clinic series, no tumor of 1 cm or less recurred given subcutaneously every 4 to 8 hours to maintain the action of
after resection. Therefore, appendectomy alone is adequate treatment. somatostatin. In addition, the long-acting repeatable agent Sandostatin
With the uncommon large appendiceal carcinoid (i.e., >2 cm) or LAR (octreotide acetate for injectable suspension) has been shown to
those with mesoappendiceal invasion, a true cancer operation (e.g., be highly effective in control of both carcinoid flushing and diarrhea.418
right hemicolectomy) is required.403,404 Small rectal lesions may be Somatostatin influences the inhibition of numerous GI hormones,
resected endoscopically. However, tumors larger than 2 cm in diameter gastric secretion, gastric and small intestinal motility, splanchnic blood
require standard cancer operations, such as a low anterior resection or flow, pancreatic enzyme secretion, intestinal nutrient absorption, and
abdominoperineal resection. However, controversy still exists regarding gallbladder contractility.419 Kvols and colleagues first described the
intermediate-sized tumors (1–2 cm confined to mucosa and submucosa). use of the somatostatin analogue octreotide in therapy for carcinoid
Endoscopic submucosal dissection (ESD) has been found effective syndrome.420 Fifty-seven patients with carcinoid tumor and carcinoid
in 239 patients with rectal carcinoid smaller than 2 cm.405 In small syndrome were treated with daily doses of octreotide acetate, ranging
bowel tumors, small bowel resection with removal of mesenteric nodes from 100 to 1127 mg (mean, 414 mg). Flushing was abolished in
is recommended. It follows that the surgical approach to bronchial, most patients, and diarrhea was controlled adequately in approximately
gastric, or gonadal carcinoid will depend on the location and stage at 75%. Control of symptoms usually was associated with a decrease in
presentation. These tumors often elicit a mesenteric fibrosing reaction, the urinary 5-HIAA levels, but reduction in tumor bulk was not seen
in which the mesentery becomes shortened and the bowel kinked, consistently. Increased doses of somatostatin may partially overcome
frequently causing partial small bowel obstruction.406 Pain or physiologic resistance.421
abnormalities secondary to partial bowel obstruction may be greatly A randomized, double-blind, placebo-controlled trial of octreotide
relieved by palliative surgical resection, bypass, or both. The indolent LAR in midgut NETs showed a significant prolongation in PFS from
course of carcinoid tumors mandates a high index of suspicion for 6 months in the placebo arm to 14.3 months in the Sandostatin LAR
Cancer of the Endocrine System  •  CHAPTER 68 1099

arm.422 The improvement in PFS was most pronounced in patients embolization, chemoembolization, or radioembolization capitalizes on
with resected primary tumors and relatively low (<10% liver involve- this anatomic principle. Although the data are not as robust as they
ment) tumor burden. An effect on overall survival could not be reliably are for colorectal cancer metastases or hepatocellular carcinoma, a
determined in this population. Furthermore, in randomized phase III number of case series have shown symptomatic benefit, improvement
trials, octreotide and lanreotide have been proven to inhibit tumor in circulating biomarkers, and radiographic responses in NET. The
growth.422,423 Data from a randomized controlled study of the effect largest series to date is from the MD Anderson Cancer Center, where
of octreotide on long-term survival among patients with metastatic 69 patients with carcinoid tumors were treated with either bland
NETs showed no difference between the octreotide and placebo embolization or chemoembolization. A radiographic response was
groups.424 There remains some question as to how long therapy with seen in 46 patients (66.7%), with a median PFS of 22.7 months.439
octreotide should continue after disease progression, or if it should Other series have reported symptomatic improvement in more than
continue indefinitely for symptomatic relief. 90% of patients receiving intraarterial therapy.440 Emerging treatments
Somatostatin analogues are usually well tolerated.419,420,425 Long-term with radioactive microspheres (yttrium-90 [90Y]) have shown some
therapy may predispose to the formation of gallstones; the drug efficacy as well.441 Radiofrequency ablation also has been used, with
promotes cholelithiasis by inhibition of cholecystokinin release and documented improvement in symptoms, although the case series are not
a resultant inhibition of gallbladder emptying. Early reports suggested as large.442,443
a high rate of cholelithiasis for patients on somatostatin and recom-
mended elective cholecystectomy, when feasible.425 However, other Targeted Agents
reports have indicated that cholelithiasis is rarely symptomatic and Inhibition of mTOR demonstrated preclinical efficacy in carcinoid
that cholecystectomy can be performed selectively.426,427 tumors,444 and everolimus, an mTOR inhibitor, recently demonstrated
a survival benefit in pNET (see later).445 The multinational RADIANT-2
Chemotherapy (RAD001 in Advanced Neuroendocrine Tumors, Second Trial) study
Because the disease is indolent, the role of cytotoxic chemotherapy randomized patients to octreotide plus everolimus versus placebo. In
has been somewhat limited. Antineoplastic therapy may be called for 429 enrolled patients, an improvement in PFS was noted (16.4 months
in patients with tumors that have a high Ki-67 level or with severe versus 11.3 months; P = .026). More biochemical improvements in
symptoms of carcinoid syndrome uncontrollable by other methods. 5-HIAA and CgA levels were seen in the everolimus arm, but there
High-grade NETs should be treated with a platinum doublet akin to was no difference in radiographic response rate. Median overall survival
treatment in small cell lung cancer.428,429 had not been reached in this trial by the time of the publication of
For lower-grade tumors, cytotoxic chemotherapy is much less its findings, and the results may be confounded because patients on
effective. With single agents, the response rates of greater than 10% placebo were allowed to cross over to everolimus. The subsequent
were seen with only three adequately tested drugs: doxorubicin, 7 of RADIANT-4 trial compared everolimus alone versus placebo for
33 (21%); 5-FU, 5 of 19 (26%); and dacarbazine (DTIC), 2 of 15 patients with advanced, progressive, nonfunctional NETs of the lung
(13%).394 A larger series of patients was involved in a phase III study and GI tract. Everolimus was associated with improved PFS (11.0
by the Eastern Cooperative Oncology Group (ECOG)430 comparing versus 3.9 months; P < .0001) and overall survival (hazard ratio [HR]
5-FU plus doxorubicin (FA) to 5-FU plus streptozocin (FS). Response = 0.64; P = .04). These findings held in subgroup analyses of both
rates (33% versus 26%, respectively) and survival for the two treatment lung and GI tumors.446 In February 2016, the FDA approved everolimus
arms were not significantly different. A subsequent ECOG trial for the treatment of adults with progressive, locally advanced, well-
comparing dose-modified FS and doxorubicin alone also demonstrated differentiated, nonfunctional NETs of GI tract origin. Stomatitis,
equivalent response rates. In the largest randomized ECOG trial of rash, and fatigue were the most common adverse events associated
208 patients, FA and FS therapies were associated with response rates with everolimus.447
of 13% and 16%, respectively; there was a trend toward improved The addition of bevacizumab to octreotide showed a response rate
survival in the FS group (24 months versus 16 months for FA; of 18% in a phase II study, with improvement in PFS when compared
P = .11).431 with interferon.448 Larger-scale studies have not yet been performed.
Sunitinib has shown efficacy in phase III studies of pNET,449 but
Interferon limited response rates were seen in a carcinoid subset when examined
Interferon has historical significance as a possible treatment for carcinoid in a phase II setting.450 More studies exploring potential targets for
tumors, but its toxicity and relative lack of efficacy limit its use. In carcinoid tumors are urgently sought.
early trials, response rates ranged from 20% to 55%, with response
gauged by decrease in symptom burden rather than radiographic Radionuclide Therapy
response.432,433 Little radiographic response has been shown with Research in radiopharmaceuticals has yielded an emerging treatment
interferon. Patients with carcinoid tumors treated with interferon may option for tumors that overexpress somatostatin receptors. Coupling
develop a wide variety of autoimmune diseases, such as thyroid disease receptor-binding somatostatin analogues (DOTATOC for octreotide,
(thyrotoxicosis, hypothyroidism), pernicious anemia, and vasculitis. DOTATATE for octreotate) with 90Y or lutetium-177 (177Lu) has
Accounting for these toxicities, the lack of significant benefit, and the been explored in clinical trials. 90Y appears to have greater efficacy
presence of more effective therapies, the use of interferon for carcinoid in larger tumors, and 177Lu in smaller tumors. Phase II data suggest
tumors is not common. Biochemical response was reported to be a partial response rate of 23% by RECIST (Response Evaluation
around 75% in patients treated with a combination of interferon and Criteria in Solid Tumors) for 90Y DOTATATE,451 and 30% for
octreotide after failure of octreotide alone.434,435 As a result, both ENETS 177
Lu DOTATATE.452 More than 50% of patients in these studies
and the North American Neuroendocrine Tumor Society (NANETS) experienced a clinical response in terms of symptom improve-
guidelines include interferon-α as a second-line therapy for functioning ment.451,452 Combining 90Y and 177Lu, a cohort study was performed
GI NETs after failure of a somatostatin analogue.436,437 in Switzerland that demonstrated an overall survival of 5.51 years.
Preliminary results of the NETTER-1 trial, in which 230 patients with
Hepatic-Directed Therapy inoperable somatostatin receptor–positive midgut NET progressive
Surgical metastasectomy for liver tumors is ideal, but possible in disease in whom standard octreotide therapy had failed, showed that
only 10% to 20% of patients with metastatic disease.438 Carcinoid 177
Lu DOTATATE was associated with improved PFS compared with
tumors have a propensity to metastasize to the liver, and derive a octreotide alone.453 Toxicities of these agents include myelosuppression,
significant component of their blood supply preferentially from and a 10% rate of severe, potentially life-threatening renal toxic-
hepatic arterial vasculature. Targeting the hepatic arterial supply with ity.454 These agents are not available in the United States outside of a
1100 Part III: Specific Malignancies

clinical trial at this time but are growing in popularity where offered part, nonautonomous. This is thought to be a result of tumor cell
in Europe. regulation by somatostatin and its predominant tumor cell recep-
tor somatostatin receptor subtype 2.456 In evaluating patients with
PANCREATIC NEUROENDOCRINE TUMORS pNET, it is important to understand that there appear to be two
different etiologies. The first group consists of those patients who
The advent of radioimmunoassay (RIA), immunofluorescence, and experienced their tumors stochastically, in the absence of significant
other techniques for identifying peptide hormones has expanded our personal or family history of endocrine disorders. The second group
knowledge of the prevalence and endocrine effects of pNETs. A includes those with clear evidence of an inherited predisposition
population-based study has shown an increased incidence of NETs to multiple neoplasias of the endocrine system in an autosomal
from 2.4% to 5.8% per 100,000 per year.455 dominant pattern. The latter patients tend to have a more indolent
The pancreatic islet contains α-cells (glucagon), β-cells (insulin), course than those with sporadic tumors. These MEN syndromes were
δ-cells (somatostatin), and enterochromaffin cells (serotonin). These described earlier. Body mass index, diabetes, smoking, and alcohol
cells are all part of the neuroendocrine system, and tumors so derived consumption have been described as risk factors for pNET in two
secrete a wide variety of polypeptides. Some of these peptides share meta-analyses.457,458 As with carcinoid tumors, the approach to the
the characteristics and functions of classic hormones: (1) their release patient with a pNET must be individualized, balancing management
follows a physiologic stimulus; (2) they have the ability to affect of the effects of hormone production with symptoms of tumor bulk.
response in a distant organ; and (3) these effects are mimicked by In any individual patient, either management issue may predominate.
exogenous infusion of the hormone. By contrast, some abnormal Treatment should be directed not only by the presence of symptoms,
peptides produced by islet cell tumors have no known clinical but also by a consideration of the relatively lengthy natural history of
hormonal effects. These peptide-secreting tumors are, for the most pNET (Fig. 68.9).

Symptom/finding

Pancreatic mass Hypoglycemia Gastric ulceration Glucose intolerance Severe watery


No systemic syndrome (diaphoresis, ± diarrhea diarrhea
seizure, coma) Hypokalemia
achlorhydria
Basal acid output With dermatitis Cholelithiasis (WDHA)
Carcinoid >15 mEq/h (migratory ± steatorrhea
Measure Nonfunctioning Fasting plasma necrolytic
pancreatic islet cell (>60% gastriN >1,000 erythema)
polypeptide malignant) pg/mL
or
+ secretin test Glucagon Elevated Pancreatic tumor
+ calcium infusion levels usually somatostatin ↑ VIP
test high
Ppoma Provocative
(>60% test rarely
Gastrinoma necessary Somatostatinoma VIPoma
malignant) (60%–90% malignant) (80% malignant (80% malignant
60% pancreatic) 90% pancreatic)

Duodenum Pancreatic
Suspect insulinoma (20%) (40%–50%)

Glucagonoma
(60% malignant
Glucose < 50 mg/dl Hypoglycemia + Serum >99% pancreatic)
Elevated plasma ↑ insulin sulfonylurea
insulin levels
↓ proinsulin
Elevated proinsulin ↓ c-peptide
Elevated or normal + insulin
c-peptide antibodies

Insulinoma Surreptitious Surreptitious oral


(10%–15% insulin use hypoglycemia use
malignant >99%
pancreatic)

Figure 68.9  •  Diagnosis of pancreatic islet cell tumors. Ppoma, Pancreatic polypeptide-secreting tumor; VIP, vasoactive intestinal peptide; VIPoma, vasoactive
intestinal peptide secreted by a pancreatic islet tumor.
Cancer of the Endocrine System  •  CHAPTER 68 1101

Diagnosis and Imaging human chorionic gonadotropin.476,477 The typical patient with symptoms
of insulinoma has a single small, pancreatic nodule. In atypical cases,
Specific syndromes and diagnostic tests for each of the more common with multiple primary endocrine tumors, including an insulinoma,
pNETs are discussed separately later. However, a few generally applicable MEN1 syndrome should be suspected.478 Insulinomas are found with
principles should be understood. Where possible, tissue confirmation equal proclivity throughout the pancreas.479
with biopsy demonstrating characteristic neuroendocrine staining These tumors usually are imaged with high-resolution multiphasic
(discussed earlier) is critical. RIA of peptides obtained by selective CT, MRI, or abdominal ultrasonography as the initial study. Occult
venous catheterization may be helpful in localizing tumors and may lesions can be identified with techniques such as endoscopic ultrasound
demonstrate the presence of metastatic spread, particularly in patients and selective arteriography and portal venous sampling.469 If these
with gastrinomas that are occult on imaging (Zollinger-Ellison syn- studies are negative, some authors recommend proceeding with
drome). NSE or CgA is useful in detecting and following therapeutic exploration with manual palpation of the pancreas and intraoperative
response in pNET, but there are limitations in diagnostic specificity, ultrasound examination, citing success rates greater than 90% at initial
mentioned earlier, with regard to carcinoid tumors.363,364 exploration.
For larger tumors, CT scans are particularly helpful in tumor Surgical resection usually is curative because of the small size and
localization and assessment of metastatic burden. CT scan can reveal low metastatic potential of insulinomas. The type of resection indicated
lesions as small as 4 mm with sensitivity greater than 80%; however, depends on the size and anatomic relationship of the tumor to the
sensitivity decreases with tumor less than 2 mm.459,460 Because many surrounding vasculature. Laparoscopic surgery can be done for small,
pNETs and their metastases may be small, the procedure may not be solitary tumors that have been localized preoperatively.480 Enucleation
adequately sensitive, and multimodality imaging may be required.461 is possible, but for larger tumors, a formal pancreatectomy is necessary.
As with most endocrine neoplasms, pNETs have a rich vascularity, As in other NETs, partial resection may afford palliation in patients
so characteristic enhancement patterns on multiphase CT and MRI whose symptoms are disabling or cannot be controlled with nonsurgical
may helpful in differentiating these tumors from adenocarcinoma. modalities, with long-term remission rates of 77% to 100%.481
Improvements in technology and technique have improved the A patient with unresectable insulinoma and recurrent episodes of
diagnostic usefulness of MRI. pNETs typically have high signal intensity hypoglycemia often benefits from appropriate diet and administration
on T2-weighted and fat-saturated images, coupled with low intensity of an insulin antagonist. Frequent feedings between meals and at
on T1-weighted and fat-saturated images, and arterial enhancement bedtime are administered with sufficient glucose to control symptoms.
with gadolinium contrast. This constellation of imaging findings has Adjustments in the carbohydrate content of the diet may be required,
a sensitivity of 94% for intrapancreatic lesions.462–465 depending on the reactivity of the individual tumor, because the
Ultrasonography is of some use in imaging both pancreatic primary stimulus of a large glucose load may lead to an exaggerated release of
tumors and hepatic metastases and may be used to guide percutane- insulin.482 Parenteral glucose supplementation becomes an important
ous biopsy of suspicious liver lesions. Endoscopic ultrasonography is adjunct in frequent or sustained hypoglycemic attacks; in emergencies,
somewhat operator dependent but may allow for image-guided FNA rapid injection of 50% glucose can be required.
for tissue confirmation of tumors.466 In a prospective series of 37 Octreotide is valuable in the general management of all hormone-
patients, endoscopic ultrasonography was highly sensitive and specific producing NETs and has been found to reduce plasma insulin levels
for pancreatic endocrine tumors, as ultimately confirmed with surgical in at least 65% of patients with insulinoma.483 Octreotide is especially
excision.467 68Ga-DOTATATE PET-CT has been shown to be effective useful when there are insulinoma metastases, because there appears
in identifying the primary lesion in patients with metastatic disease.385 to be upregulation of somatostatin receptor subtype 2 of the metastatic
lesions (whereas as few as 17% of the primary lesions express this
Staging receptor).479 Initiation of octreotide acetate requires close monitoring
when used in insulinoma therapy, because up to 60% of primary
Like the carcinoid subset of NET, staging guidelines for pNET have tumors may not have SST2 receptors, and hypoglycemia may worsen.484
been suggested that offer greater prognostic potential than the staging Corticosteroids, human growth hormone, and glucagon have been
for pancreatic adenocarcinoma (see Table 68.12). useful palliative agents in individual patients.485,486 However, because
of their limited effectiveness, they are best used in combination with
Specific Pancreatic Neuroendocrine other antihormonal measures. Furthermore, glucagon stimulates
Tumor Subtypes pancreatic insulin secretion and may cause paradoxical exacerbation
of a hypoglycemic episode.
Insulinoma Diazoxide’s potent hyperglycemic properties, originally recognized
Insulinomas are the most common functional pNET.468 The average during its use as an antihypertensive agent, have now been extended
age of presentation of insulinoma is in the fifth decade, with a 57% to the palliation of insulinoma and leucine-sensitive hypoglycemia of
female preponderance.469 An incidence of 4 cases per million per year infancy.487 Its principal action is to inhibit insulin release directly from
has been reported.470 The sine qua non in diagnosing this syndrome the β-cell. It also may have an extrapancreatic hyperglycemic effect
is fasting or inappropriate hypoglycemia accompanied by a relatively and cause marked edema and hirsutism. Diazoxide is administered
high plasma insulin level.471,472 Other tests have been proposed, orally in divided doses, ranging from 100 to 1000 mg/day. Although
including a hypernormal response to tolbutamide, the response of the plasma insulin level often can be reduced to a level that causes
plasma insulin to an infusion of calcium gluconate, and the ratio of no symptoms, the tumor will continue to grow and metastasize if
proinsulin471,473 to insulin in the plasma, which occasionally is not malignant, because diazoxide lacks anticancer activity. Insulinomas
useful because the absolute insulin level is not elevated in all insulino- have excellent long-term outcomes; 20-year survival has been reported
mas. In addition, measurement of β-hydroxybutyrate is helpful in in 90% of patients with metastatic insulinomas.488
diagnosing insulinomas owing to the antiketogenic effect of insulin.
The key to the diagnosis is a high index of suspicion. Glucagonoma
Sporadic insulinomas usually are single, smaller than 2 cm, and Glucagon from pancreatic α-cells plays an important role in modulating
benign; approximately 10% are malignant with metastatic distribution serum glucose concentrations. Unregulated secretion of glucagon by
into lymph node, liver, and bone.474,475 Differentiation of benign from α-cell tumors produces a distinctive clinical syndrome. An annual
malignant tumors is difficult based on the pathologic features alone, incidence rate of 0.01 to 0.1 new cases per 100,000 population per
but the presence of metastases defines malignancy. In patients with year has been reported.489 A cutaneous rash, described as a necrotizing
malignant disease, proinsulin may be increased in plasma or circulating migratory erythema, is the most characteristic feature and is present
1102 Part III: Specific Malignancies

in approximately 70% of patients.490,491 Mild insulin-resistant diabetes pancreatic or duodenal wall G cells are responsible for the signs and
and weight loss attributable to the catabolic effects of glucagon also symptoms of Zollinger-Ellison syndrome, a disorder characterized by
are seen. Glossitis, cheilosis, and venous thromboses can develop. hypersecretion of gastrin. First described in 1955, this syndrome is
Glucagon inhibits intestinal motility, and the glucagonoma syndrome characterized by hypersecretion of gastric acid, severe peptic ulcer
often includes ileus and constipation. Because symptoms commonly disease, and an islet cell tumor of the pancreas. It is estimated that
are mild and nonspecific, the tumor often is recognized late, when less than 0.1% of patients with peptic ulcer disease have Zollinger-
metastases are present.491 Most tumors have grown to larger than 4 cm Ellison syndrome.326 Eighty percent of gastrinomas are sporadic, and
at diagnosis, and 50% to 80% are metastatic.492 Most glucagonomas the rest occur in association with MEN1.507
are sporadic; only 3% are associated with MEN1, and fewer are linked The hallmark of the gastrinoma syndrome is recurrent peptic ulcer
with von Hippel-Lindau syndrome.493,494 disease in spite of adequate medical or surgical treatment. Intermittent
The diagnostic test for glucagonoma is the finding of a high plasma diarrhea, often with steatorrhea, may be present as a result of digestive
glucagon concentration (normal, <60 pg/mL). In patients with glu- enzyme inactivation in the small intestine by unbuffered gastric acid.
cagonoma, the plasma hormone level typically is markedly elevated All manifestations of the Zollinger-Ellison syndrome are secondary
and often is greater than 1000 pg/mL. The diagnosis is further suggested to hypersecretion of gastric acid. A history of MEN1 syndrome has
by failure of glucose to suppress glucagon, by an abnormal rise in great significance, and gastrinoma may be present in up to 50% of
plasma glucagon following infusion of arginine, by the presence of these patients.508 The combination of high gastric acid secretion and
hypoaminoacidemia, and, if tumor is available, immunoperoxidase hypergastrinemia is strongly suggestive of gastrinoma, but this combina-
staining for glucagon. Reviews have suggested that the tumor is more tion also can occur in patients with retained gastric antrum after
common in women and typically manifests in the fifth and sixth surgery for peptic ulcer (antrectomy and Billroth II gastric resection)
decades.492,495 Symptoms persist for many years before the diagnosis and after gastric outlet obstruction. Gastric rugal hypertrophy, multiple
is made; survival, even with metastatic disease, may be lengthy. The ulcers, or ulceration of the small bowel on radiographic studies suggests
primary tumor is in the tail or body of the pancreas in 50% of patients gastrinoma.509
and in the head of the pancreas in 8% of patients; the remaining 42% In a review of 60 patients treated surgically for gastrinoma at Ohio
of patients have diffuse involvement. CT scans and MRI are useful State University, Ellison and associates reported that the incidence of
in identification of the primary tumor, as is somatostatin receptor MEN was 27%, and a primary tumor was detected in nearly 90% of
scintigraphy (SSRS) because most are larger than 3 cm at presenta- cases. Twenty-five percent of gastrinomas can occur in the pancreas.
tion.496,497 Presence of calcification in CT indicates lymph node Extrapancreatic locations include the duodenum, predominantly the
metastasis.498 The tumor is resectable for cure in less than one-third first part of the duodenum, and the stomach, retroperitoneal lymph
of cases, and recurrence after resection, mainly in the liver, is common. nodes, liver, and bile duct.489,510 More than one-third of patients in
However, liver resection is appropriate for metastatic disease.499 In the Ohio State series had multiple tumors; metastatic disease in the
addition to surgical resection, octreotide produces an improvement liver was identified in 20% of patients.511
of skin rash in up to 90% of patients and complete disappearance of The clinical diagnosis of Zollinger-Ellison syndrome has changed.
rash in 30% with symptomatic control of glucagon hypersecretion.500 Although the original case reports stressed the appearance of extensive
Chemotherapeutic agents may have some activity (see later). The and multiple gastric ulcers, a heightened index of suspicion and early
tyrosine kinase inhibitor sunitinib has shown good efficacy in one detection have altered this pattern of disease presentation. Suspicion
clinical trial, with prolongation of survival for up to 11.4 months.445 should be raised in any patient with peptic ulcer disease refractory to
In spite of the overwhelming malignant presentation of these tumors, Helicobacter pylori eradication. Classically, the diagnosis of Zollinger-
the overall 5-year survival rate of 50% without resection is quite Ellison syndrome is based on four steps. The first step is to identify
encouraging and is likely due to slow progression.501 fasting hypergastrinemia in association with a basal acid output greater
than 15 mEq/h. In general, a gastrin level greater than 10 times the
Somatostatinoma upper limit of normal (1000 pg/mL) in the presence of gastric PH
Somatostatin first was identified in pituitary cells, and a role in the below 2 is pathognomonic. Less-convincing elevations in fasting serum
regulation of growth hormone secretion was ascribed to it. Subsequently, gastrin can be further evaluated with the secretin test, in which a peak
it was recognized as a hormone of the islet δ-cells. Somatostatin may level of serum gastrin higher than 200 pg/mL over the baseline after
serve as a paracrine regulator of other pancreatic islet cell hormones. administration of secretin is considered diagnostic.508,512 A calcium
Inhibition of secretion of those hormones may account for some of test was recommended after a prospective study as a second-line test
the signs of somatostatinoma, such as diabetes, diarrhea, or gallbladder in patients in whom suspicion of gastrinoma is high but the result of
disorders.502 secretin test is negative.513 The remaining three steps include documenta-
Somatostatinoma occurs most frequently in the head of the pancreas, tion of peptic ulcer disease (usually with endoscopy), localization of
and as many as 80% of patients have evidence of metastases at diagnosis. the primary tumor, and assessment of malignancy. Approximately
The mean age at presentation is 55 years.503 Between 70% and 92% 75% of patients with Zollinger-Ellison syndrome have solitary ulcers
of somatostatinomas are metastatic at presentation.504 Somatostatinomas smaller than 1 cm in the first part of the duodenum, 14% in the
produce such common symptoms as diabetes (type 2–like diabetes distal part, and 11% in the jejunum.514 Important to note, widespread
mellitus) and gallbladder disease; therefore the clinician usually does use of PPIs makes biochemical diagnosis of gastrinoma more chal-
not consider the diagnosis of somatostatinoma until late in the disease, lenging, because PPIs raise both gastrin levels and CgA levels. A fasting
by which time metastases are likely. Surgical resection usually is not specimen, off PPI therapy for 10 to 14 days, is ideal but should not
curative.502 Because of the relatively mild hormonally induced symptoms be considered in the setting of uncontrolled peptic ulcer disease with
produced by somatostatin, octreotide does not have the same palliative bleeding.515
benefit it has in other pNETs. However, somatostatin analogues are Localization of gastrinomas has been discussed extensively. Tech-
considered the first line of treatment of unresectable tumors.505 niques such as endoscopic ultrasonography, CT scan, MRI, selective
Cytoreductive surgery and chemotherapy may be the most appropriate venous sampling for gastrin, and abdominal arteriography all have a
palliative strategies. The overall 5-year survival rate is 75%.506 Metastatic role in the diagnosis and management of this disease. As in other
disease is associated with 5-year survival of 60%. NETs, octreotide scanning is useful for identifying primary and
metastatic lesions.516 Because gastrinomas are so frequently metastatic,
Gastrinoma it is necessary to use every diagnostic modality to rule out metastatic
Gastrin, the polypeptide hormone normally secreted by the G cell of disease before planning surgery. If no metastatic disease is identified,
the gastric antrum, stimulates gastric acid secretion. Tumors of the potentially curable operative intervention is indicated.517,518
Cancer of the Endocrine System  •  CHAPTER 68 1103

Medical management in gastrinoma is directed toward the hyper- prolonged PFS among patients with metastatic enteropancreatic
secretion of gastric acid. Before the introduction of histamine receptor NETs of grade 1 or 2 in a double-blind placebo-randomized trial.423
antagonists, the only practical way to treat recurrent duodenal and The short duration of response and low incidence of objective tumor
jejunal ulcers was total gastrectomy. PPIs may completely eradicate regression suggest that somatostatin analogues in the treatment of
symptoms of peptic ulcer disease caused by acid hypersecretion. Two metastatic islet cell tumors have a more limited role than in other GI
series using PPIs illustrated that symptomatic relief can be achieved in carcinoid tumors.
more than 90% of patients with PPIs alone, with safe escalation of PPI
doses titrated to achieve gastric acid levels of less than 10 mEq/h.519,520 Interferon
Higher doses are necessary initially but can be titrated down steadily As with carcinoid tumors, interferon has historical significance in the
over time as patients tolerate the PPI. In refractory cases, octreotide treatment of pNETs. Swedish investigators reported on 22 patients
may be considered. Octreotide can be used if PPIs are unable to control treated with human leukocyte interferon, with an objective response
gastric acid secretion. Surgical resection of sporadic gastrinomas with rate of 77% and a median duration of response of 8.5 months.432
no evidence of metastatic disease is associated with a 50% to 97% Most of these responses were documented by decreased hormone
long-term survival rate.521,522,522a production rather than radiographic response. Clinical trials still
investigate the use of interferon, particularly in pegylated form, but
Tumors Secreting Vasoactive Intestinal Peptide its use outside of clinical trials is not recommended.529
In 1958, Verner and Morrison described a syndrome of watery diarrhea,
hypokalemia, hypochlorhydria, and metabolic acidosis (WDHA Chemotherapy
syndrome), which is caused by high circulating levels of VIP secreted Cytotoxic chemotherapy for pNET is not a first choice for therapy.
by a pNET (VIPoma).523 Most studies of VIP infusions in healthy Chemotherapy usually is attempted in patients with symptoms caused
volunteers have supported the concept that the diarrhea in VIPoma by tumor bulk that may not be palliated by cytoreductive surgery
patients may be caused directly by elevated circulating levels of vasoactive or in patients with uncontrolled syndromes of hormone excess. In
intestinal polypeptide.524 Because most patients with this syndrome contrast to carcinoid tumors, pNETs generally are more responsive
have metastatic disease at presentation, usually to the liver, management to chemotherapy. The first chemotherapeutic drug to elicit significant
is mainly medical, with chemotherapy or a somatostatin analogue. attention in the treatment of pNET was the antitumor antibiotic
Surgical resection is rarely curative. As with carcinoid tumors and streptozotocin. This drug has a diabetogenic action in some animals
other islet cell tumors, however, in patients with locally unresectable that is correlated with selective uptake of the drug by pancreatic
disease or those with hepatic metastases, surgical cytoreduction or β-cells.530 An ECOG study from 1980 compared streptozotocin alone
regional therapy may improve symptom control. These tumors are and streptozotocin plus 5-FU in advanced islet cell carcinomas.531 The
exceedingly rare and are associated with metastases in the chest or combination was superior to streptozotocin alone in overall rate of
retroperitoneum.525 response (63% versus 36%). These responses generally were of long
duration and yielded meaningful improvements in performance status
Therapy and symptoms, but were not reproduced in later studies in which more
standardized radiographic response criteria were used.532 More recent
Radiation Therapy studies in the RECIST era have demonstrated response rates of 39%
Radiotherapy has similar indications as carcinoid tumors for symp- to 53% with streptozocin-based therapy, combined with either 5-FU
tomatic lesions, particularly in the late and rare complication of bony or doxorubicin or both.533–535 The combined results of two phase II
metastatic disease. Among patients who are not candidates for surgical trials, although both studies have not met their primary end point,
resection, some data from case reports and small case series indicate demonstrated radiologic response and prolonged disease stability in
that radiation therapy can produce symptomatic improvement and pNETs treated with oxaliplatin-fluoropyrimidine chemotherapy plus
freedom from local progression.526,527 bevacizumab.536
Dacarbazine (DTIC) has shown efficacy in pNET, and in newer
Liver-Directed Therapy studies has been replaced by orally available temozolomide. Combina-
As with carcinoid tumor, islet cell tumors often result in predominantly tions of temozolomide plus thalidomide, or temozolomide plus
hepatic metastases, and reduction in tumor bulk in the liver may capecitabine have demonstrated promising results in phase II studies,
significantly influence hormone production and quality of life. For with radiographic response rates as high as 70%.537,538 This combination
that reason, resection of hepatic metastases is warranted in selected offers a possible cytotoxic combination for further study.
patients. Endocrine and tumor response to hepatic arterial occlu-
sion, with or without chemotherapy, can be impressive. The Mayo Targeted Therapy
Clinic experience documented 46 patients with islet cell tumor Inhibition of the mTOR pathway demonstrated potential efficacy in
regression rates of 43% with hepatic artery occlusion alone versus survival prolongation in preclinical and early phase studies, even without
78.1% when chemotherapy was included. In the MD Anderson frequent radiographic responses.445 In the phase III RADIANT-3 study,
series, 54 patients had pNETs, with a response rate of 35.2%, a everolimus (10 mg/day) was compared with placebo and demonstrated
median PFS of 16.1 months, and a median overall survival of an improvement in PFS from 4.6 months to 11.0 months, with 34%
23.2 months.439 of patients alive without progression at 18 months of therapy. This
result was achieved with the modest radiographic response rate of 5%
Somatostatin Analogue (2% for placebo).445 Patients were allowed to cross over at progression,
The somatostatin analogue octreotide is as useful in the treatment which may have confounded the overall survival data, but the improve-
of syndromes associated with ectopic hormone production in islet ment in PFS is compelling for everolimus. In preliminary report from
cell tumors as it is in treatment of carcinoid tumors. In a series from a phase II trial that enrolled 150 patients with locally advanced or
the Mayo Clinic, 24 patients with islet cell tumor were treated. The metastatic pNETs, everolimus alone or in combination with bevaci-
response to somatostatin analogue was prompt, and symptoms were zumab, the combination group was associated with a significantly
palliated. However, the median duration of response was only 2.5 higher response rate (31% versus 12%) and superior PFS (16.7%
months, with only 2 of 24 patients (8%) continuing to benefit beyond versus 14%).539
1 year.528 In another review of 66 patients treated with octreotide, At the same time, sunitinib has been used in pNET with some
only 8 patients (12%) showed any indication of objective tumor success in clinical trials. In phase II studies, a response rate of 17%
response.483 However, lanreotide was associated with significant with a significant rate of disease stabilization was identified, prompting
1104 Part III: Specific Malignancies

a move toward phase III studies.450 The findings of the phase III over many years. Sporadic (nonfamilial) parathyroid carcinomas
study were published in 2011 and demonstrated an improvement in frequently carry HRPT2 mutations, too. In two different studies,
PFS from 5.5 months with placebo to 11.4 months with sunitinib. HRPT2 mutations were reported to occur in 10 of 15547 and in 4 of
Similarly, the objective response rate was low (9%), and overall 4 sporadic parathyroid carcinomas.548 Most mutations were somatic
survival data may be confounded because of early stoppage of the as opposed to germline in HPT-JT. Unsuspected germline mutations,
trial.449 A phase III trial demonstrated improved PFS with sunitinib however, were also discovered in a minority of patients with clinical
compared with placebo (11.4 versus 5.5 months).449 The develop- presentation of sporadic disease, suggesting that some of these individu-
ment of these two new compounds with significant improvement als may have phenotypic variants of HPT-JT.548 The recognition that
in PFS marks a breakthrough in medical therapy for pNET. These family members of some patients with apparently sporadic parathyroid
therapies may usher in a new era of therapy for metastatic NET, as carcinomas are also at risk for parathyroid malignancy has created a
two well-tolerated, orally available compounds have been proven to new indication for genetic testing (see section on genetic counseling,
prolong PFS. later). An obvious question is how prevalent are inactivating mutations
of parafibromin in benign parathyroid adenomas. In the largest series
of sporadic benign parathyroid adenomas reported to date, no mutations
PARATHYROID CARCINOMA of the HRPT2 gene could be detected.549 However, prevalence of
Incidence HRPT2 mutation was reported as 1.8% in two different studies.546,550
Other genes, through somatic mutation, also appear to participate in
Parathyroid carcinoma is a malignant growth of parathyroid chief the pathogenesis of parathyroid carcinoma. As examples, evidence has
cells. Severe hyperparathyroidism is the most common form of presenta- implicated abnormal expression of the retinoblastoma and p53 pro-
tion and it represents less than 1% of patients with primary hyper- teins.551,552 In another report, somatic mutations in the MEN1 gene
parathyroidism.540,541 It is one of the rarest cancers, with an estimated were found in 3 of 23 sporadic parathyroid carcinoma cases,553 although
prevalence of 0.005%.540,542,543 Histologically, abnormal parathyroid diagnostic criteria for carcinoma in this study differed from other
glands from patients with hyperparathyroidism have been characterized studies in not requiring local invasion or metastases. No such mutations
as being hyperplastic, adenomatous, or malignant. Single-gland had previously been reported in parathyroid carcinoma. Furthermore,
involvement (“adenoma”) occurs in approximately 80% of patients parathyroid carcinoma is not considered to be part of the MEN1-
with hyperparathyroidism, and multiple-gland involvement (“hyper- associated phenotype. In a series of 348 cases of MEN1, only one
plasia”) in approximately 20%. Less than 2% of hyperfunctioning case (0.28%) of parathyroid carcinoma was reported.554
parathyroid glands are malignant. The SEER cancer registry data
identified 224 patients with parathyroid carcinoma between 1998 Clinical Characteristics
and 2003 in the United States.7 During this time period, the incidence
of parathyroid carcinoma increased from 3.58 to 5.73 per 10 million Symptoms of parathyroid carcinoma are primarily caused by high
people. An incidence rate of 0.36 between 2000 and 2012 was reported levels of calcium in serum (hypercalcemia). Summarized from four
in a SEER registry recent publication.544 Whereas benign hyperpara- studies,547,555,556 they include:
thyroidism is more common in women (3 : 1), the incidence of
• Constitutional symptoms (32%–74%):
parathyroid carcinoma is equal between the two sexes. The age of
• Muscle weakness, fatigue
diagnosis is reportedly between 44 and 54 years.7
• Nausea, vomiting, anorexia, constipation
• Increased thirst, frequent urination
Etiology • Bone pain, fractures (34%–73%)
• Neck mass (34%–52%)
As in many other cancers, a single-cell mutational event is believed
• Kidney stones (32%–70%)
to give rise to parathyroid neoplasms. Whereas monoclonal tumors
• Pancreatitis (0%–15%)
likely result from somatic mutations in one parent cell (adenoma and
• No symptoms (2%–7%)
carcinoma), polyclonal expansion is thought to develop when multiple
original cells (hyperplasia) respond to an exogenous or endogenous Signs of parathyroid carcinoma are also a consequence of hyper-
stimulus. Thus, monoclonal parathyroid carcinomas are likely caused parathyroidism, as follows:
by single-cell derangement from mutation in either growth-promoting
• Elevated serum calcium (14.6–15.9 mg/dL; >14 mg/dL in 70%)
or growth-regulating genes.
• Elevated serum PTH (commonly 10-fold higher than the upper
Studies have demonstrated that a subset of parathyroid adenomas
limit of normal)
harbor a specific rearrangement of chromosome 11—inv(11)
(p15;q13)—a pericentromeric inversion that positions the PTH gene’s Although most patients with parathyroid carcinoma have hyper-
5′ regulatory region upstream of the cyclin D1 gene, thereby driving calcemia, a few patients are normocalcemic with a neck mass. In
overexpression of cyclin D1 with the consequent growth promotion.545 contrast to benign parathyroid adenomas, 40% to 70% of parathyroid
As for tumor suppressor genes in parathyroid carcinoma, chromosome cancers are palpable on physical examination.542,556,557 Nonfunctioning
1q25 harvests HRPT2 (hereditary hyperparathyroidism type 2) gene, parathyroid carcinomas are extremely rare, with fewer than 20 cases
a gene responsible for growth control. Inactivating mutation(s) of this reported.558,559 In contrast to patients with functional parathyroid
tumor suppressor gene (also called cell division cycle 73 [CDC73]) carcinomas, patients with nonfunctional tumors die from mass effect
has been recognized to play a central role in the molecular pathogenesis and tumor burden, rather than from hypercalcemia.558 Multiglandular
of many parathyroid carcinomas. HRPT2 encodes parafibromin, a (still monoclonal) parathyroid carcinoma is also extremely rare.560
protein whose function appears to involve regulation of gene expression Lymph node metastases have been observed in up to one-third of
and inhibition of cell proliferation. Inactivated germline mutations patients with parathyroid carcinoma. Another one-third of patients
in the HRPT2 gene are responsible for an autosomal dominant type have metastasis to other sites, including lung, liver, and bone.561
of familial hyperparathyroidism, the hyperparathyroidism–jaw tumor
syndrome (HPT-JT).546 Patients with HPT-JT are predisposed to Diagnosis
develop ossifying fibromas of the jaw, cystic and neoplastic renal lesions,
uterine tumors, and parathyroid neoplasia with an increased risk of Because the clinical and biochemical presentation of hyperparathyroid-
parathyroid carcinoma (15%). In HPT-JT all parathyroid glands are ism is common to benign and malignant diseases, diagnosis of para-
at risk for tumor development, but the tumors can occur asynchronously thyroid carcinoma is usually established after the first surgery. Ideally,
Cancer of the Endocrine System  •  CHAPTER 68 1105

preoperative confirmation or suspicion for parathyroid carcinoma would capsular disruption and tumor spillage during surgery. If the diagnosis
lead to more aggressive surgical excision and therefore a higher cure of parathyroid carcinoma is made postoperatively, several authors
rate at the time of first surgery. A high index of suspicious should recommend reoperation with ipsilateral thyroidectomy,579 but a survival
exist in patients with marked hypercalcemia (serum calcium ≥14 mg/ benefit has not been clearly demonstrated.561,580 If during surgery, a
dL), very high PTH (3–10 times above the upper limit of normal for parathyroid tumor appears to be well demarcated (not invading sur-
the assay), or profound hypercalcemic symptoms with simultaneous rounding tissue) but is later found on pathologic examination to have
manifestations of renal and bone involvement.557,562,563 For localization invaded adjacent tissue, reoperation to remove all tumor tissue is
purposes, patients should undergo a special radioactive scan of the recommended. After surgical resection, recurrence rates of up to 49%
parathyroid glands (technetium-99m sestamibi scan). A neck ultrasound to 60% have been reported.541,542,556,561,580
examination with or without FNA washout for PTH measurement Surgical complications such as hematoma, recurrent laryngeal nerve
may also help in localizing parathyroid tumors. Sonographic features injury, injury to surrounding structures, and postoperative metabolic
that suggest parathyroid carcinoma include heterogeneous or lobulated complications including hypocalcemia and hypophosphatemia have
lesions and large size.564,565 These tests are done to confirm which been reported, with a perioperative mortality rate of 1.8%.542,563,580
parathyroid gland is abnormal but do not discriminate benign from The postoperative management of patients with parathyroid carcinoma
malignant disease. Because of the difficulty in diagnosing these tumors, should include close monitoring of the patient’s serum calcium
they usually are at a more advanced stage at presentation.558,566 concentration. Patients with parathyroid carcinoma are especially at
Other diagnostic imaging modalities that could be beneficial for risk of developing “hungry bone syndrome” and therefore need to be
disease localization and metastatic or recurrent disease include selective preventively treated with large doses of calcium and calcitriol. Patients
venous sampling, CT scan, and MRI, with sensitivity of 83%, 93%, with recurrent or metastatic parathyroid carcinoma can be treated
and 69%, respectively.542 surgically. Resection of lesions in the neck, lungs, and liver will often
Owing to difficulty in differentiating benign from malignant disease result in significant palliation of hypercalcemia.542
and the possibility of tumor rupture and seeding, FNA is not recom-
mended in preoperative diagnosis of parathyroid carcinoma.567 Medical Therapy
Once the specimen has been removed, pathologic examination The following treatments may be used to correct hypercalcemia caused
should be done to determine malignancy. The histopathologic examina- by parathyroid carcinoma preoperatively and when the condition is
tion of parathyroid carcinoma may show uniform sheets of cells arranged no longer amenable to surgery:
in a lobular pattern separated by dense fibrous trabeculae, mitotic
• Drugs that acutely lower serum calcium level (e.g., gallium nitrate),
figures in the tumor cells, and capsular and blood vessel invasion.568,569
calcitonin (hypocalcemic hormone), and hydration (intravenous
However, these features have been noted occasionally in parathyroid
fluids) and loop diuretics can be used as first-line treatments.
adenomas. Gross invasion beyond the capsule, including extracapsular
• Calcimimetics are drugs that reduce PTH secretion by producing
invasion, appears to correlate best with subsequent tumor recurrence.
allosteric changes in the calcium-sensing receptor that induce signal-
Thus, because of the nonspecific nature of these histologic features,
ing without the need of calcium or at lower thresholds.581 Cinacalcet,
the two criteria with which a definitive diagnosis of parathyroid
a longer-acting calcimimetic drug, is used for the treatment of
carcinoma can be made are:
secondary hyperparathyroidism associated with renal failure and
• Local invasion of contiguous structures and/or hypercalcemia associated with parathyroid carcinoma. In a 16-week
• Lymph node or distant metastases open-label study of 29 patients with inoperable parathyroid carci-
noma, cinacalcet (dose titrated to achieve calcium ≤10 mg/dL or
In some cases, it may not be possible to differentiate parathyroid
up to 90 mg four times daily) successfully reduced serum calcium
adenoma from carcinoma at the time of diagnosis or initial surgery.
concentration by at least 1 mg/dL in 62% of patients.581 Mean
Local recurrence or the occurrence of distal metastases at subsequent
PTH levels decreased but not significantly. Adverse events (nausea,
follow-up ultimately determines the correct pathologic diagnosis.
vomiting, headache, dehydration) were common and resulted in
Some authors have suggested a benefit of immunohistochemical
discontinuation in five patients.
panels that include parafibromin, galactin-3, PGD9.5, and Ki-67 in
• The bisphosphonate pamidronate has been reported to improve
diagnosing parathyroid carcinoma, with a sensitivity of 80% and
hypercalcemia in individual cases of parathyroid carcinoma.582 A
specificity of 100%.570
similar response was reported for zoledronic acid, which is more
The HPRT2 gene is inactive in approximately 67% of sporadic
potent than pamidronate in treating hypercalcemia of malignancy.583
parathyroid cancers.547 However, there is no single molecular marker
• Denosumab, an osteoblast inhibitor that binds to the receptor
that can distinguish between parathyroid carcinoma and adenoma.571
activator of nuclear factor–κB ligand (RANKL), has been suc-
Some literature has suggested that an inverted third-generation to
cessfully used to treat refractory hypercalcemia in some case
second-generation PTH ratio is a tumor marker that could dif-
reports.584–587
ferentiate patients with parathyroid carcinomas among patients
with primary hyperparathyroidism, with sensitivity of 78.5% and
specificity of 98.9%.572–575 Adjuvant Therapy
Both adjuvant chemotherapy and radiation therapy have had generally
Treatment poor results for the treatment of parathyroid carcinoma. Use of either
should be considered as a last resource—only when a patient is a poor
Surgical Therapy candidate for surgery and/or hypercalcemia cannot be controlled.
Total parathyroidectomy or en bloc resection of the parathyroid mass Radiation therapy experience is limited to small observational
with negative margins and any adjacent tissues that have been invaded studies. In a series of 26 patients with locally invasive disease followed
by tumor is currently the treatment of choice.576 In the SEER registry, for a mean of 7.9 years (range, 2 to 21 years), only 1 of 6 patients
78.6% of patients underwent total parathyroidectomy and 12.5% receiving adjuvant radiation therapy had a local relapse, compared
underwent en bloc resection.7 En bloc resection could include the with 10 of 20 who had not received radiation therapy.561
ipsilateral thyroid lobe, paratracheal alveolar and lymphatic tissue, the Chemotherapy has been tried whenever other treatment modalities
thymus, or some of the neck muscles, and in some advanced cases have failed. Various agents, alone or in combination, have resulted in
the recurrent laryngeal nerve.555,556,577 Some surgeons recommend occasional responses. An example of one patient with pulmonary
ipsilateral lymph node dissection.578 Because parathyroid carcinoma metastases responding to treatment with dacarbazine, 5-FU, and
can be seeded locally from the primary tumor, it is important to avoid cyclophosphamide with normalization of serum calcium for 13 months
1106 Part III: Specific Malignancies

has been reported, but there are no randomized controlled trials assessing regarding surveillance in these families. Thus, genetic testing for
the efficacy of these agents.563 germline HRPT2 mutation is clinically appropriate in most patients
Biologic agents based on gene products such as recombinant with sporadic parathyroid carcinoma.
parafibromin, an inhibitor of cell proliferation in parathyroid neoplasia,
telomerase inhibitors such as azidothymidine, and immune therapy Outcomes
constitute novel emerging therapies with encouraging in vitro results
and may prove useful clinically in the future.576 Parathyroid carcinomas grow slowly in most patients but can occasion-
Radiofrequency ablation of metastatic disease with unresectable ally be aggressive. The disease typically seems to follow one of three
disseminated lesions has been reported, sometimes in combination courses: one-third of patients are cured at initial or follow-up surgery;
with transcatheter arterial embolization.588,589 one-third develop recurrence after a prolonged disease-free period but
Finally, in metastatic parathyroid carcinoma with hypercalcemia, may be cured with reoperation; and one-third of patients experience
immunotherapy with PTH-like immunogenic fragments for PTH a short and aggressive course.542 Survival, however, may be improving.
antibody production has shown some promising results in terms of The National Cancer Database survey (1985–1995) reported 5- and
improving hypercalcemia and quality of life.572,590,591 10-year survival rates of 55.5% and 49%, respectively, in a series of
286 patients.593 In a study using the SEER cancer registry (1988–2003),
Genetic Counseling 5- and 10-year survival rates were 83.9% and 67.8%, respectively.543
Genetic HRPT2 testing is indicated when features suggestive of HPT-JT These survival rates are consistent with a 2016 report from the SEER
are present in the index patient. Genetic diagnosis of a germline cancer registry, with a 5-year survival rate of 82.5% and a 10-year
HRPT2 inactivating mutation indicates that such a patient may have survival rate of 64.8%.544 The recurrence rate is 50% or greater among
classic HPT-JT, expressing its initial manifestation, or phenotypic patients with lymph node or distant metastasis, with a mean time of
variants, such as familial isolated hyperparathyroidism, or a form of recurrence ranging between 2.5 and 4.8 years.561,580 In functioning
HPT-JT with altered penetrance. A subset of patients with sporadic parathyroid carcinoma, recurrence usually manifests as elevated serum
parathyroid carcinoma have been found to have germline HRPT2 calcium and/or PTH. Young age, female sex, recent year of diagnosis,
mutations,547 and such mutations carry important implications for and absence of distant metastases were associated with improved
management of the patient and/or for early detection and prevention survival.
of parathyroid malignancy in family members. A negative test result,
however, is a challenging situation because among families with classic The complete reference list is available online at
HPT-JT, 40% have no detectable mutation.550,592 There is no consensus ExpertConsult.com.

KEY REFERENCES
5. Haugen BR, Alexander EK, Bible KC, Doherty GM, for patients younger than age 45 years with papillary analysis of 700 patients. J Clin Endocrinol Metab.
Mandel SJ, Nikiforov YE, et al. 2015 American thyroid cancer. J Clin Oncol. 2015;33:2370– 1997;82:3553–3562.
Thyroid Association management guidelines for 2375. 137. Wada N, Duh QY, Sugino K. Lymph node metas-
adult patients with thyroid nodules and differentiated 72. Edge SB, Byrd DR, Compton CC, et al (eds). AJCC tasis from 259 papillary thyroid microcarcinomas:
thyroid cancer: the American Thyroid Association Cancer Staging Handbook: From the AJCC Cancer frequency, pattern of occurrence and recurrence,
Guidelines Task Force on Thyroid Nodules and Dif- Staging Manuel. 7th ed. New York: Springer-Verlag; and optimal strategy for neck dissection. Ann Surg.
ferentiated Thyroid Cancer. Thyroid. 2016;26:1–133. 2010. 2003;237:399–407.
10. Zarebczan B, Chen H. Multi-targeted approach 77. Chen H, Nicol TL, Zeiger MA. Hürthle cell 147. Tuttle RM, Ball DW, Byrd D. Thyroid carcinoma.
in the treatment of thyroid cancer. Minerva Chir. neoplasms of the thyroid: are there factors predictive J Natl Compr Canc Netw. 2010;8:1228–1274.
2010;65:59–69. of malignancy? Ann Surg. 1998;227:542–546. 156. Sawka AM, Brierley JD, Tsang RW, Thabane L,
14. Fraker DL. Radiation exposure and other factors 92. Mazzaferri EL. Management of a solitary thyroid Rotstein L, Gafni A, et al. An updated systematic
that predispose to human thyroid neoplasia. Surg nodule. N Engl J Med. 1993;328:553–559. review and commentary examining the effective-
Clin North Am. 1995;75:365–375. 97. American Thyroid Association (ATA) Guidelines ness of radioactive iodine remnant ablation in
21. Nikiforova MN, Lynch RA, Biddinger PW. RAS Taskforce on Thyroid Nodules and Differentiated well-differentiated thyroid cancer. Endocrinol Metab
point mutations and PAX8-PPAR gamma rear- Thyroid Cancer, Cooper DS, Doherty GM, Haugen Clin North Am. 2008;37:457–480, x.
rangement in thyroid tumors: evidence for distinct BR, et al. Revised American Thyroid Association 171. Brierley JD, Tsang RW. External-beam radiation
molecular pathways in thyroid follicular carcinoma. management guidelines for patients with thyroid therapy in the treatment of differentiated thyroid
J Clin Endocrinol Metab. 2003;88:2318–2326. nodules and differentiated thyroid. cancer. Thyroid. cancer. Semin Surg Oncol. 1999;16:42–49.
28. Fagin JA, Mitsiades N. Molecular pathology of 2009;19:1167–11214. 178. Brose MS, Nutting CM, Jarzab B, Elisei R, Siena
thyroid cancer: diagnostic and clinical implica- 100. Brito JP, Ito Y, Miyauchi A, Tuttle RM. A clinical S, Bastholt L, et al. Sorafenib in radioactive iodine-
tions. Best Pract Res Clin Endocrinol Metab. framework to facilitate risk stratification when refractory, locally advanced or metastatic differenti-
2008;22:955–969. considering an active surveillance alternative to ated thyroid cancer: a randomised, double-blind,
54. Mazeh H, Chen H. Advances in surgical therapy immediate biopsy and surgery in papillary micro- phase 3 trial. Lancet. 2014;384:319–328.
for thyroid cancer. Nat Rev Endocrinol. 2011;7:581– carcinoma. Thyroid. 2016;26:144–149. 192. Roman S, Lin R, Sosa JA. Prognosis of medullary
588. 105. Cha YJ, Koo JS. Next-generation sequencing in thyroid carcinoma: demographic, clinical, and
55. Lang BH, Chow SM, Lo CY. Staging systems for thyroid cancer. J Transl Med. 2016;14:322. pathologic predictors of survival in 1252 cases.
papillary thyroid carcinoma: a study of 2 tertiary 106. Nikiforov YE. Thyroid carcinoma: molecular Cancer. 2006;107:2134–2142.
referral centers. Ann Surg. 2007;246:114–121. pathways and therapeutic targets. Mod Pathol. 199. Sippel RS, Kunnimalaiyaan M, Chen H. Current
56. DeGroot LJ, Kaplan EL, McCormick M, Straus 2008;21(suppl 2):S37–S43. management of medullary thyroid cancer. Oncologist.
FH. Natural history, treatment, and course of papil- 112. Sippel RS, Chen H. Controversies in the surgical 2008;13:539–547.
lary thyroid carcinoma. J Clin Endocrinol Metab. management of newly diagnosed and recurrent/ 200. Chen H, Sippel RS, O’Dorisio MS, Vinik AI,
1990;71:414–424. residual thyroid cancer. Thyroid. 2009;19:1373–1380. Lloyd RV, Pacak K. The North American Neuro-
64. Podnos YD, Smith D, Wagman LD, Ellenhorn JD. 131. Mazzaferri EL, Young RL. Papillary thyroid car- endocrine Tumor Society consensus guideline for
The implication of lymph node metastasis on survival cinoma: a 10 year follow-up report of the impact the diagnosis and management of neuroendocrine
in patients with well-differentiated thyroid cancer. of therapy in 576 patients. Am J Surg. 1981;70: tumors: pheochromocytoma, paraganglioma, and
Am Surg. 2005;71:731–734. 511–518. medullary thyroid cancer. Pancreas. 2010;39:775–
65. Adam MA, Pura J, Goffredo P, Dinan MA, Reed SD, 133. Loh KC, Greenspan FS, Gee L. Pathological 783.
Scheri RP, et al. Presence and Number of lymph node tumor-node-metastasis (pTNM) staging for papillary 220. Ogilvie JB, Kebebew E. Indication and timing
metastases are associated with compromised survival and follicular thyroid carcinomas: a retrospective of thyroid surgery for patients with hereditary
Cancer of the Endocrine System  •  CHAPTER 68 1107

medullary thyroid cancer syndromes. J Natl Compr with adrenocortical carcinoma. Ann Surg Oncol. 397. Roberts WC, Sjoerdsma A. The cardiac disease
Canc Netw. 2006;4:139–147. 1999;6:719–726. associated with the carcinoid syndrome (carcinoid
221. Moley JF, DeBenedetti MK. Patterns of nodal 288. Zarnegar R, Kebebew E, Duh QY, Clark OH. heart disease). Am J Med. 1964;36:5–34.
metastases in palpable medullary thyroid carcinoma: Malignant pheochromocytoma. Surg Oncol Clin 407. Kim MK, Warner RR, Roayaie S, Harpaz N,
recommendations for extent of node dissection. Ann N Am. 2006;15:555–571. Ward SC, Itzkowitz S, et al. Revised staging clas-
Surg. 1999;229:880–887, discussion 887–888. 291. Neumann HP, Bausch B, McWhinney SR. Germ-line sification improves outcome prediction for small
227. Chen H, Roberts JR, Ball DW. Effective long-term mutations in nonsyndromic pheochromocytoma. N intestinal neuroendocrine tumors. J Clin Oncol.
palliation of symptomatic, incurable metastatic Engl J Med. 2002;346:1459–1466. 2013;31:3776–3781.
medullary thyroid cancer by operative resection. 302. Guller U, Turek J, Eubanks S. Detecting pheochro- 422. Rinke A, Muller HH, Schade-Brittinger C. Promid,
Ann Surg. 1998;227:887–895. mocytoma: defining the most sensitive test. Ann J. Placebo-controlled, double-blind, prospective,
231. de Groot JW, Zonnenberg BA, van Ufford-Mannesse Surg. 2006;243:102–107. randomized study on the effect of octreotide LAR
PQ. A phase II trial of imatinib therapy for meta- 305. Greenblatt DY, Shenker Y, Chen H. The utility of in the control of tumor growth in patients with
static medullary thyroid carcinoma. J Clin Endocrinol metaiodobenzylguanidine (MIBG) scintigraphy in metastatic neuroendocrine midgut tumors: a report
Metab. 2007;92:3466–3469. patients with pheochromocytoma. Ann Surg Oncol. from the Group. J Clin Oncol. 2009;27:4656–4663.
253. Young WF Jr. Clinical practice. The inciden- 2008;15:900–905. 445. Yao JC, Shah MH, Ito T. Everolimus for advanced
tally discovered adrenal mass. N Engl J Med. 307. Strong VE, Kennedy T, Al-Ahmadie H. Prognostic pancreatic neuroendocrine tumors. N Engl J Med.
2007;356:601–610. indicators of malignancy in adrenal pheochromocyto- 2011;364:514–523.
258. Young WF Jr. Conventional imaging in adrenocorti- mas: clinical, histopathologic, and cell cycle/apoptosis 446. Yao JC, Fazio N, Singh S, Buzzoni R, Carnaghi
cal carcinoma: update and perspectives. Horm Cancer. gene expression analysis. Surgery. 2008;143:759–768. C, Wolin E, et al. Everolimus for the treatment of
2011;2:341–347. 308. Thompson LD. Pheochromocytoma of the Adrenal advanced, non-functional neuroendocrine tumours
270. Terzolo M, Angeli A, Fassnacht M. Adjuvant gland Scaled Score (PASS) to separate benign from of the lung or gastrointestinal tract (RADIANT-4):
mitotane treatment for adrenocortical carcinoma. malignant neoplasms: a clinicopathologic and a randomised, placebo-controlled, phase 3 study.
N Engl J Med. 2007;356:2372–2380. immunophenotypic study of 100 cases. Am J Clin Lancet. 2016;387:968–977.
271. Grubbs EG, Callender GG, Xing Y. Recurrence Pathol. 2002;26:551–566. 447. Pavel ME, Hainsworth JD, Baudin E. Everolimus
of adrenal cortical carcinoma following resection: 357. Yip L, Cote GJ, Shapiro SE. Multiple endocrine neo- plus octreotide long-acting repeatable for the
surgery alone can achieve results equal to surgery plasia type 2: evaluation of the genotype-phenotype treatment of advanced neuroendocrine tumours
plus mitotane. Ann Surg Oncol. 2010;17:263– relationship. Arch Surg. 2003;138:409–416, discus- associated with carcinoid syndrome (RADIANT-2):
270. sion 416. a randomised, placebo-controlled, phase 3 study.
275. Fassnacht M, Terzolo M, Allolio B, et al. Combi- 361. Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Lancet. 2011;378:2005–2012.
nation chemotherapy in advanced adrenocortical Mares JE, et al. One hundred years after “carcinoid”: 449. Raymond E, Dahan L, Raoul JL, et al. Sunitinib
carcinoma. N Engl J Med. 2012;366:2189–2197. epidemiology of and prognostic factors for neuro- malate for the treatment of pancreatic neuroendo-
283. Fassnacht M, Berruti A, Baudin E, Demeure MJ, endocrine tumors in 35,825 cases in the United crine tumors. N Engl J Med. 2011;364:501–513.
Gilbert J, Haak H, et al. Linsitinib (OSI-906) States. J Clin Oncol. 2008;26:3063–3072. 551. Cryns VL, Thor A, Xu HJ, et al. Loss of the reti-
versus placebo for patients with locally advanced 380. Reidy-Lagunes DL, Gollub MJ, Saltz LB. J. Addition noblastoma tumor-suppressor gene in parathyroid
or metastatic adrenocortical carcinoma: a double- of octreotide functional imaging to cross-sectional carcinoma. N Engl J Med. 1994;330:757–761.
blind, randomised, phase 3 study. Lancet Oncol. computed tomography or magnetic resonance 561. Busaidy NL, Jimenez C, Habra MA. Parathyroid
2015;16:426–435. imaging for the detection of neuroendocrine carcinoma: a 22-year experience. Head Neck.
284. Schulick RD, Brennan MF. Long-term survival after tumors: added value or an anachronism? J Clin 2004;26:716–726.
complete resection and repeat resection in patients Oncol. 2011;29:e74–e75.
Cancer of the Endocrine System  •  CHAPTER 68 1107.e1
1107.e1

REFERENCES
1. Patel KN, Shaha AR. Poorly differentiated thyroid cancer. J Clin Endocrinol Metab. 2013;98: polyposis: molecular analysis of pathogenesis in
and anaplastic thyroid cancer. Cancer Control. E1852–E1860. a family and review of the literature. Endocr J.
2006;13:119–128. 21. Nikiforova MN, Lynch RA, Biddinger PW. RAS 2004;51:317–323.
2. Udelsman R, Chen H. The current management point mutations and PAX8-PPAR gamma rear- 38. Loh KC. Familial nonmedullary thyroid carcinoma: a
of thyroid cancer. Adv Surg. 1999;33:1–27. rangement in thyroid tumors: evidence for distinct meta-review of case series. Thyroid. 1997;7:107–113.
3. Davies L, Welch HG. Current thyroid cancer trends molecular pathways in thyroid follicular carcinoma. 39. Malchoff CD, Malchoff DM. Familial non-
in the United States. JAMA Otolaryngol Head Neck J Clin Endocrinol Metab. 2003;88:2318–2326. medullary thyroid carcinoma. Cancer Control.
Surg. 2014;140:317–322. 22. Nikiforov YE, Nikiforova MN. Molecular genetics 2006;13:106–110.
4. Leenhardt L, Bernier MO, Boin-Pineau MH, and diagnosis of thyroid cancer. Nat Rev Endocrinol. 40. Sippel RS, Caron NR, Clark OH. An evidence-based
Conte Devolx B, Marechaud R, Niccoli-Sire P, 2011;7:569–580. approach to familial nonmedullary thyroid cancer:
et al. Advances in diagnostic practices affect thyroid 23. Niemeier LA, Kuffner Akatsu H, Song C, screening, clinical management, and follow-up. World
cancer incidence in France. Eur J Endocrinol. Carty SE, Hodak SP, Yip L, et al. A combined J Surg. 2007;31:924–933.
2004;150:133–139. molecular-pathologic score improves risk stratifica- 41. Bonora E, Tallini G, Romeo G. Genetic predisposition
5. Haugen BR, Alexander EK, Bible KC, Doherty GM, tion of thyroid papillary microcarcinoma. Cancer. to familial nonmedullary thyroid cancer: an update
Mandel SJ, Nikiforov YE, et al. 2015 American 2012;118:2069–2077. of molecular findings and state-of-the-art studies.
Thyroid Association management guidelines for 24. Zheng X, Wei S, Han Y, Li Y, Yu Y, Yun X, et al. J Oncol. 2010;2010:385206.
adult patients with thyroid nodules and differentiated Papillary microcarcinoma of the thyroid: clinical 42. Bevan S, Pal T, Greenberg CR. A comprehensive
thyroid cancer: the American Thyroid Association characteristics and BRAF(V600E) mutational status analysis of MNG1, TCO1, fPTC, PTEN, TSHR,
Guidelines Task Force on Thyroid Nodules and Dif- of 977 cases. Ann Surg Oncol. 2013;20:2266–2273. and TRKA in familial nonmedullary thyroid cancer:
ferentiated Thyroid Cancer. Thyroid. 2016;26:1–133. 25. Lin KL, Wang OC, Zhang XH, Dai XX, Hu XQ, confirmation of linkage to TCO1. J Clin Endocrinol
6. Brito JP, Al Nofal A, Montori VM, Hay ID, Morris Qu JM. The BRAF mutation is predictive of aggres- Metab. 2001;86:3701–3704.
JC. The impact of subclinical disease and mechanism sive clinicopathological characteristics in papillary 43. Bignell GR, Canzian F, Shayeghi M. Familial
of detection on the rise in thyroid cancer incidence: thyroid microcarcinoma. Ann Surg Oncol. 2010;17: nontoxic multinodular thyroid goiter locus maps
a population-based study in Olmsted County, 3294–3300. to chromosome 14q but does not account for familial
Minnesota During 1935 through 2012. Thyroid. 26. Medema RH, Bos JL. The role of p21ras in nonmedullary thyroid cancer. Am J Hum Genet.
2015;25:999–1007. receptor tyrosine kinase signaling. Crit Rev Oncog. 1997;61:1123–1130.
7. Ries LAG, Melbert D, Krapcho M, Bethesda MD. 1993;4:615–661. 44. McKay JD, Lesueur F, Jonard L. Localization of a
SEER cancer statistics review; 2007: 1975-2005. 27. Garcia-Rostan G, Zhao H, Camp RL, Pollan M, susceptibility gene for familial nonmedullary thyroid
National Cancer Institute. Herrero A, Pardo J, et al. ras mutations are associated carcinoma to chromosome 2q21. Am J Hum Genet.
8. Davies L, Welch HG. Increasing incidence of with aggressive tumor phenotypes and poor prognosis 2001;69:440–446.
thyroid cancer in the United States. JAMA. in thyroid cancer. J Clin Oncol. 2003;21:3226–3235. 45. Wang X, Cheng W, Li J, Su A, Wei T, Liu F, et al.
2006;295:1973–2002. 2164-7. 28. Fagin JA, Mitsiades N. Molecular pathology of Endocrine tumours: familial nonmedullary thyroid
9. Moo-Young TA, Traugott AL, Moley JF. Sporadic thyroid cancer: diagnostic and clinical implications. carcinoma is a more aggressive disease: a systematic
and familial medullary thyroid carcinoma: state of Best Pract Res Clin Endocrinol Metab. 2008;22: review and meta-analysis. Eur J Endocrinol. 2015;172:
the art. Surg Clin North Am. 2009;89:1193–1204. 955–969. R253–R262.
10. Zarebczan B, Chen H. Multi-targeted approach in the 29. Gianoukakis AG, Giannelli SM, Salameh WA, 46. Zhang Q, Yang S, Meng XY, Chen G, Pang RZ.
treatment of thyroid cancer. Minerva Chir. 2010;65: McPhaul LW. Well differentiated follicular thyroid Clinical analysis of familial nonmedullary thyroid
59–69. neoplasia: impact of molecular and technological carcinoma. World J Surg. 2016;40:570–573.
11. Tuttle RM, Becker DV. The Chernobyl accident advances on detection, monitoring and treatment. 47. Mazeh H, Benavidez J, Poehls JL. In patients with
and its consequences: update at the millennium. Mol Cell Endocrinol. 2011;332:9–20. thyroid cancer of follicular cell origin, a family
Semin Nucl Med. 2000;30:133–140. 30. Ohori NP, Schoedel KE. Variability in the atypia history of nonmedullary thyroid cancer in one
12. Cetta F, Montalto G, Petracci M, Fusco A. Thyroid of undetermined significance/follicular lesion first-degree relative is associated with more aggressive
cancer and the Chernobyl accident. Are longterm of undetermined significance diagnosis in the disease. Thyroid. 2012;22:3–8.
and long distance side effects of fallout radiation Bethesda System for Reporting Thyroid Cytopa- 48. Bradly DP, Reddy V, Prinz RA, Guttuso P. Incidental
greater than estimated Endocrinol Metab. J Clin thology: sources and recommendations. Acta Cytol. papillary carcinoma in patients treated surgically
Endocrinol Metab. 1997;82:2015–2017. 2011;55:492–498. for benign thyroid diseases. Surgery. 2009;146:
13. Hancock SL, Cox RS, McDougall IR. Thyroid 31. Cantara S, Capezzone M, Marchisotta S, Capuano S, 1099–1104.
diseases after treatment of Hodgkin’s disease. N Busonero G, Toti P, et al. Impact of proto-oncogene 49. From G, Mellemgaard A, Knudsen N. Review of
Engl J Med. 1991;325:599–605. mutation detection in cytological specimens from thyroid cancer cases among patients with previous
14. Fraker DL. Radiation exposure and other factors thyroid nodules improves the diagnostic accuracy benign thyroid disorders. Thyroid. 2000;10:697–700.
that predispose to human thyroid neoplasia. Surg of cytology. J Clin Endocrinol Metab. 2010;95: 50. Roti E, et al. Clinical and histological characteristics
Clin North Am. 1995;75:365–375. 1365–1369. of papillary thyroid microcarcinoma results of a
15. Horn-Ross PL, Morris JS, Lee M. Iodine and thyroid 32. Nikiforov YE, Ohori NP, Hodak SP, Carty SE, retrospective study in 243 patients. J Clin Endocrinol
cancer risk among women in a multiethnic popula- LeBeau SO, Ferris RL, et al. Impact of mutational Metab. 2006;91:2171–2178.
tion: the Bay Area Thyroid Cancer Study. Cancer testing on the diagnosis and management of patients 51. Mazzaferri EL. Management of low-risk differentiated
Epidemiol Biomarkers Prev. 2001;10:979–985. with cytologically indeterminate thyroid nodules: a thyroid cancer. Endocr Pract. 2007;13:498–512.
16. Knobel M, Medeiros-Neto G. Relevance of prospective analysis of 1056 FNA samples. J Clin 52. Roti E, degli Uberti EC, Bondanelli M, Braverman
iodine intake as a reputed predisposing factor for Endocrinol Metab. 2011;96:3390–3397. LE. Thyroid papillary microcarcinoma: a descriptive
thyroid cancer. Arq Bras Endocrinol Metabol. 2007; 33. Nikiforov YE, Steward DL, Robinson-Smith TM, and meta-analysis study. Eur J Endocrinol. 2008;159:
51:701–712. Haugen BR, Klopper JP, Zhu Z, et al. Molecular 659–673.
17. Alberti L, Carniti C, Miranda C. RET and NTRK1 testing for mutations in improving the fine-needle 53. Yu XM, Wan Y, Sippel RS, Chen H. Should all papil-
proto-oncogenes in human diseases. J Cell Physiol. aspiration diagnosis of thyroid nodules. J Clin lary thyroid microcarcinomas be aggressively treated?
2003;195:168–186. Endocrinol Metab. 2009;94:2092–2098. An analysis of 18,445 cases. Ann Surg. 2011;254:
18. Fusco A, Chiappetta G, Hui P. Assessment of 34. Beaudenon-Huibregtse S, Alexander EK, Guttler RB, 653–660.
RET/PTC oncogene activation and clonality in Hershman JM, Babu V, Blevins TC, et al. Central- 54. Mazeh H, Chen H. Advances in surgical therapy for
thyroid nodules with incomplete morphological ized molecular testing for oncogenic gene mutations thyroid cancer. Nat Rev Endocrinol. 2011;7:581–588.
evidence of papillary carcinoma: a search for the complements the local cytopathologic diagnosis of 55. Lang BH, Chow SM, Lo CY. Staging systems for
early precursors of papillary cancer. Am J Pathol. thyroid nodules. Thyroid. 2014;24:1479–1487. papillary thyroid carcinoma: a study of 2 tertiary
2002;160:2157–2167. 35. Integrated genomic characterization of papillary referral centers. Ann Surg. 2007;246:114–121.
19. Fugazzola L, Puxeddu E, Avenia N. Correlation thyroid carcinoma. Cell. 2014;159:676–690. 56. DeGroot LJ, Kaplan EL, McCormick M, Straus
between B-RAFV600E mutation and clinico-patho- 36. Liu X, Qu S, Liu R, Sheng C, Shi X, Zhu G, FH. Natural history, treatment, and course of papil-
logic parameters in papillary thyroid carcinoma: data et al. TERT promoter mutations and their associa- lary thyroid carcinoma. J Clin Endocrinol Metab.
from a multicentric Italian study and review of the tion with BRAF V600E mutation and aggressive 1990;71:414–424.
literature. Endocr Relat Cancer. 2006;13:455–464. clinicopathological characteristics of thyroid cancer. 57. Solorzano CC, Carneiro DM, Ramirez M, Lee TM,
20. Nikiforova MN, Wald AI, Roy S, Durso MB, J Clin Endocrinol Metab. 2014;99:E1130–E1136. Irvin GL 3rd. Surgeon-performed ultrasound in
Nikiforov YE. Targeted next-generation sequenc- 37. Lee S, Hong SW, Shin SJ. Papillary thyroid the management of thyroid malignancy. Am Surg.
ing panel (ThyroSeq) for detection of mutations in carcinoma associated with familial adenomatous 2004;70:576–580, discussion 80-82.
1107.e2 Part III: Specific Malignancies

58. Shimamoto K, Satake H, Sawaki A, Ishigaki T, of the thyroid gland with a review of the English 97. Doherty GM, Haugen BR, Kloos RT. Revised Ameri-
Funahashi H, Imai T. Preoperative staging of literature. Cancer. 2001;91:505–524. can Thyroid Association management guidelines for
thyroid papillary carcinoma with ultrasonography. 77. Chen H, Nicol TL, Zeiger MA. Hürthle cell patients with thyroid nodules and differentiated
Eur J Radiol. 1998;29:4–10. neoplasms of the thyroid: are there factors predictive thyroid cancer. Thyroid. 2009;19:1167–1214.
59. Qubain SW, Nakano S, Baba M, Takao S, Aikou of malignancy? Ann Surg. 1998;227:542–546. 98. Mazzaferri EL, Robbins RJ, Spencer CA. A con-
T. Distribution of lymph node micrometastasis in 78. Harness JK, Thompson NW, McLeod MK. Follicular sensus report of the role of serum thyroglobulin
pN0 well-differentiated thyroid carcinoma. Surgery. carcinoma of the thyroid gland: trends and treatment. as a monitoring method for low-risk patients with
2002;131:249–256. Surgery. 1984;96:972–980. papillary thyroid carcinoma. J Clin Endocrinol Metab.
60. Arturi F, Russo D, Giuffrida D, Ippolito A, Per- 79. Phitayakorn R, McHenry CR. Follicular and Hürthle 2003;88:1433–1441.
rotti N, Vigneri R, et al. Early diagnosis by genetic cell carcinoma of the thyroid gland. Surg Oncol Clin. 99. Gharib H, Papini E, Valcavi R. American Associa-
analysis of differentiated thyroid cancer metastases 2006;15:603–623, ixx. tion of Clinical Endocrinologists and Associazione
in small lymph nodes. J Clin Endocrinol Metab. 80. Zhang YW, Greenblatt DY, Repplinger D. Older age Medici Endocrinologi medical guidelines for clinical
1997;82:1638–1641. and larger tumor size predict malignancy in Hürthle practice for the diagnosis and management of thyroid
61. Wada N, Suganuma N, Nakayama H. Microscopic cell neoplasms of the thyroid. Ann Surg Oncol. 2008; nodules. Endocr Pract. 2006;12:63–102.
regional lymph node status in papillary thyroid carci- 15:2842–2846. 100. Brito JP, Ito Y, Miyauchi A, Tuttle RM. A clinical
noma with and without lymphadenopathy and its rela- 81. Wreesmann VB, Ghossein RA, Patel SG. Genome- framework to facilitate risk stratification when
tion to outcomes. Langenbecks Arch Surg. 2007;392: wide appraisal of thyroid cancer progression. Am J considering an active surveillance alternative to
417–422. Pathol. 2002;161:1549–1556. immediate biopsy and surgery in papillary micro-
62. Ito Y, Tomoda C, Uruno T. Preoperative ultraso- 82. Burman KD, Ringel MD, Wartofsky L. Unusual carcinoma. Thyroid. 2016;26:144–149.
nographic examination for lymph node metastasis: types of thyroid neoplasms. Endocrinol Metab Clin 101. Crippa S, Mazzucchelli L, Cibas ES, Ali SZ.
usefulness when designing lymph node dissection for North Am. 1996;25:49–68. The Bethesda System for reporting thyroid fine-
papillary microcarcinoma of the thyroid. WORLD. 83. Wang W, Macapinlac H, Larson SM. [18F]-2-fluoro- needle aspiration specimens. Am J Clin Pathol.
2004;28:498–501. 2-deoxy-D-glucose positron emission tomography 2010;134:343–344, author reply 345.
63. Lundgren CI, Hall P, Dickman PW, Zedenius J. localizes residual thyroid cancer in patients with 102. Vriens MR, Sabanci U, Epstein HD. Reliability
Clinically significant prognostic factors for differenti- negative diagnostic (131)I whole body scans and of fine-needle aspiration in patients with familial
ated thyroid carcinoma: a population-based, nested elevated serum thyroglobulin levels. J Clin Endocrinol nonmedullary thyroid cancer. Thyroid. 1999;9:
case-control study. Cancer. 2006;106:524–531. Metab. 1999;84:2291–2302. 1011–1016.
64. Podnos YD, Smith D, Wagman LD, Ellenhorn JD. 84. Widder S, Pasieka JL. Primary thyroid lymphomas. 103. Alexander EK, Heering JP, Benson CB, Frates
The implication of lymph node metastasis on survival Curr Treat Options Oncol. 2004;5:307–313. MC, Doubilet PM, Cibas ES, et al. Assessment
in patients with well-differentiated thyroid cancer. 85. Curtis RE, Rowlings PA, Deeg HJ, Shriner DA, Socie of nondiagnostic ultrasound-guided fine needle
Am Surg. 2005;71:731–734. G, Travis LB, et al. Solid cancers after bone marrow aspirations of thyroid nodules. J Clin Endocrinol
65. Adam MA, Pura J, Goffredo P, Dinan MA, Reed SD, transplantation. N Engl J Med. 1997;336:897–904. Metab. 2002;87:4924–4927.
Scheri RP, et al. Presence and number of lymph node 86. Steele SR, Martin MJ, Mullenix PS, Azarow KS, 104. Cameci C, Jeffrey RB, McDougall IR. Ultrasound-
metastases are associated with compromised survival Andersen CA. The significance of incidental thyroid guided fine-needle aspiration biopsy of thyroid
for patients younger than age 45 years with papillary abnormalities identified during carotid duplex masses. Thyroid. 1998;8:283–289.
thyroid cancer. J Clin Oncol. 2015;33:2370–2375. ultrasonography. Arch Surg. 2005;140:981–985. 105. Cha YJ, Koo JS. Next-generation sequencing in
66. Hay ID, Grant CS, Taylor WF, McConahey WM. 87. Hagag P, Strauss S, Weiss M. Role of ultrasound- thyroid cancer. J Transl Med. 2016;14:322.
Ipsilateral lobectomy versus bilateral lobar resec- guided fine-needle aspiration biopsy in 106. Nikiforov YE. Thyroid carcinoma: molecular pathways
tion in papillary thyroid carcinoma: a retrospective evaluation of nonpalpable thyroid nodules. Thyroid. and therapeutic targets. Mod Pathol. 2008;21(suppl 2):
analysis of surgical outcome using a novel prognostic 1998;8:989–995. S37–S43.
scoring system. Surgery. 1987;102:1088–1095. 88. Leenhardt L, Hejblum G, Franc B, Fediaevsky 107. Hodak SP, Rosenthal DS. Information for clinicians:
67. Cady B, Rossi R. An expanded view of risk-group LD, Delbot T, Le Guillouzic D, et al. Indications commercially available molecular diagnosis testing
definition in differentiated thyroid carcinoma. and limits of ultrasound-guided cytology in the in the evaluation of thyroid nodule fine-needle
Surgery. 1988;104:947–953. management of nonpalpable thyroid nodules. J aspiration specimens. Thyroid. 2013;23:131–134.
68. Hay ID, Bergstralh EJ, Goellner JR. Predicting Clin Endocrinol Metab. 1999;84:24–28. 108. Xing M, Haugen BR, Schlumberger M. Progress
outcome in papillary thyroid carcinoma: development 89. Nam-Goong IS, Kim HY, Gong G, Lee HK, Hong SJ, in molecular-based management of differentiated
of a reliable prognostic scoring system in a cohort of Kim WB, et al. Ultrasonography-guided fine-needle thyroid cancer. Lancet. 2013;381:1058–1069.
1779 patients surgically treated at one institution aspiration of thyroid incidentaloma: correlation with 109. Leenhardt L, Erdogan MF, Hegedus L, Mandel SJ,
during through. Surgery. 1993;114:1050–1057, pathological findings. Clin Endocrinol (Oxf). 2004;60: Paschke R, Rago T, et al. 2013 European Thyroid
discussion 1057–1058. 21–28. Association guidelines for cervical ultrasound scan
69. Mazzaferri EL, Jhiang SM. Long-term impact of 90. Scheumann GF, Gimm O, Wegener G. Prognostic and ultrasound-guided techniques in the postopera-
initial surgical and medical therapy on papillary and significance and surgical management of locoregional tive management of patients with thyroid cancer.
follicular thyroid cancer. Am J Med. 1994;97:418– lymph node metastases in papillary thyroid cancer. Eur Thyroid J. 2013;2:147–159.
428. WORLD. 1994;18:559–567. 110. Poehls JL, Chen H, Sippel RS. Preoperative ultra-
70. Byar DP, Green SB, Dor P. A prognostic index for 91. Belfiore A, Garofalo MR, Giuffrida D. Increased sonography findings predict the need for repeated
thyroid carcinoma. A study of the EORTC Thyroid aggressiveness of thyroid cancer in patients with surgery in papillary thyroid cancer. Endocr Pract.
Cancer Cooperative Group. Eur J Cancer. 1979;15: Graves’ disease. J Clin Endocrinol Metab. 1990;70:830– 2012;18:403–409.
1033–1041. 835. 111. Luo J, McManus C, Chen H, Sippel RS. Are
71. Sherman SI, Brierley JD, Sperling M. Prospective 92. Mazzaferri EL. Management of a solitary thyroid there predictors of malignancy in patients with
multicenter study of thyroiscarcinoma treatment: nodule. N Engl J Med. 1993;328:553–559. multinodular goiter? J Surg Res. 2012;174:207–210.
initial analysis of staging and outcome. National 93. Nagai GR, Pitts WC, Basso L. Scintigraphic hot 112. Sippel RS, Chen H. Controversies in the surgical
Thyroid Cancer Treatment Cooperative Study nodules and thyroid carcinoma. Clin Nucl Med. management of newly diagnosed and recurrent/resid-
Registry Group. Cancer. 1998;83:1012–1021. 1987;12:123–127. ual thyroid cancer. Thyroid. 2009;19:1373–1380.
72. Edge SB, Byrd DR, Compton CC, et al, eds. AJCC 94. Boelaert K, Horacek J, Holder RL, Watkinson JC, 113. Sippel RS, Elaraj DM, Poder L. Localization of recur-
Cancer Staging Handbook: From the AJCC Cancer Sheppard MC, Franklyn JA. Serum thyrotropin rent thyroid cancer using intraoperative ultrasound-
Staging Manuel. 7th ed. New York: Springer-Verlag; concentration as a novel predictor of malignancy guided dye injection. WORLD. 2009;33:434–439.
2010. in thyroid nodules investigated by fine-needle aspira- 114. Al-Azawi D, Mann GB, Judson RT, Miller JA. J.
73. Ries LAG, Eisner MP, Kosary CL, Hankey BF, Seer, tion. J Clin Endocrinol Metab. 2006;91:4295–4301. Endocrine surgeon-performed US guided thyroid
Bethesda MD Statistics Review; 2005. National 95. Haymart MR, Repplinger DJ, Leverson GE, Elson FNAC is accurate and efficient. World J Surg.
Cancer Institute. DF, Sippel RS, Jaume JC, et al. Higher serum thyroid 2012;36:1947–1952.
74. Schlumberger MJ. Papillary and follicular thyroid stimulating hormone level in thyroid nodule patients 115. Goldfarb M, Gondek SS, Lew JI. Clinic-based
carcinoma. N Engl J Med. 1998;338:297–306. is associated with greater risks of differentiated ultrasound can predict malignancy in pediatric
75. Brennan MD, Bergstralh EJ, van Heerden JA, thyroid cancer and advanced tumor stage. J Clin thyroid nodules. Thyroid. 2012;22:827–831.
McConahey WM. Follicular thyroid cancer treated Endocrinol Metab. 2008;93:809–814. 116. Kim MH, Ko SH. Role of [(18)F]-fluorodeoxy-d-
at the Mayo Clinic, through: initial manifestations, 96. Haugen BR, Ridgway EC, McLaughlin BA, glucose positron emission tomography and computed
pathologic findings, therapy, and outcome. Mayo McDermott MT. Clinical comparison of whole- tomography in the early detection of persistent/recur-
Clin Proc. 1991;66:11–22. body radioiodine scan and serum thyroglobulin after rent thyroid carcinoma in intermediate-to-high risk
76. Thompson LD, Wieneke JA, Paal E. A clinicopatho- stimulation with recombinant human thyrotropin. patients following initial radioactive iodine ablation
logic study of minimally invasive follicular carcinoma Thyroid. 2002;12:37–43. therapy. Thyroid. 2012;22:157–164.
Cancer of the Endocrine System  •  CHAPTER 68 1107.e3
1107.e3

117. Yip L, Stang MT, Carty SE. Thyroid carcinoma: 136. Eustatia-Rutten CF, Corssmit EP, Biermasz NR. 154. Chow SM, Law SC, Au SK. Differentiated thyroid
the surgeon’s perspective. Radiol Clin North Am. Survival and death causes in differentiated thyroid carcinoma: comparison between papillary and fol-
2011;49:463–471, vi. carcinoma. J Clin Endocrinol Metab. 2006;91: licular carcinoma in a single institute. Head Neck.
118. Cooper DS, Doherty GM, Haugen BR, Kloos RT, 313–319. 2002;24:670–677.
Lee SL, Mandel SJ, et al. Revised American Thyroid 137. Wada N, Duh QY, Sugino K. Lymph node metas- 155. Sacks W, Fung CH, Chang JT, Waxman A,
Association management guidelines for patients with tasis from 259 papillary thyroid microcarcinomas: Braunstein GD. The effectiveness of radioactive
thyroid nodules and differentiated thyroid cancer. frequency, pattern of occurrence and recurrence, iodine for treatment of low-risk thyroid cancer: a
Thyroid. 2009;19:1167–1214. and optimal strategy for neck dissection. Ann Surg. systematic analysis of the peer-reviewed literature
119. Matsuzu K, Sugino K, Masudo K, Nagahama M, 2003;237:399–407. from 1966 to April 2008. Thyroid. 2010;20:1235–
Kitagawa W, Shibuya H, et al. Thyroid lobectomy 138. Gemsenjager E, Perren A, Seifert B. Lymph node 1245.
for papillary thyroid cancer: long-term follow-up surgery in papillary thyroid carcinoma. J Am Coll 156. Sawka AM, Brierley JD, Tsang RW, Thabane L,
study of 1,088 cases. World J Surg. 2014;38:68–79. Surg. 2003;197:182–190. Rotstein L, Gafni A, et al. An updated systematic
120. Barney BM, Hitchcock YJ, Sharma P, Shrieve DC, 139. Bardet S, Malville E, Rame JP. Macroscopic lymph- review and commentary examining the effective-
Tward JD. Overall and cause-specific survival for node involvement and neck dissection predict lymph- ness of radioactive iodine remnant ablation in
patients undergoing lobectomy, near-total, or total node recurrence in papillary thyroid carcinoma. Eur well-differentiated thyroid cancer. Endocrinol Metab
thyroidectomy for differentiated thyroid cancer. Head J Endocrinol. 2008;158:551–560. Clin North Am. 2008;37:457–480, x.
Neck. 2011;33:645–649. 140. Roh JL, Park JY, Park CI. Total thyroidectomy plus 157. Lamartina L, Durante C, Filetti S, Cooper DS. Low-
121. Mendelsohn AH, Elashoff DA, Abemayor E, St John neck dissection in differentiated papillary thyroid risk differentiated thyroid cancer and radioiodine
MA. Surgery for papillary thyroid carcinoma: is carcinoma patients: pattern of nodal metastasis, mor- remnant ablation: a systematic review of the litera-
lobectomy enough? Arch Otolaryngol Head Neck bidity, recurrence, and postoperative levels of serum ture. J Clin Endocrinol Metab. 2015;100:1748–1761.
Surg. 2010;136:1055–1061. parathyroid hormone. Ann Surg. 2007;245:604–610. 158. Ross DS, Litofsky D, Ain KB, Bigos T, Brierley
122. Haigh PI, Urbach DR, Rotstein LE. Extent of 141. Hughes DT, Doherty GM. Central neck dissec- JD, Cooper DS, et al. Recurrence after treat-
thyroidectomy is not a major determinant of survival tion for papillary thyroid cancer. Cancer Control. ment of micropapillary thyroid cancer. Thyroid.
in low- or high-risk papillary thyroid cancer. Ann 2011;18:83–88. 2009;19:1043–1048.
Surg Oncol. 2005;12:81–89. 142. Lang BH, Wong KP, Wan KY, Lo CY. Significance 159. Kim HJ, Kim NK, Choi JH, Kim SW, Jin SM,
123. Nixon IJ, Ganly I, Patel SG, Palmer FL, Whitcher of metastatic lymph node ratio on stimulated Suh S, et al. Radioactive iodine ablation does
MM, Tuttle RM, et al. Thyroid lobectomy for treat- thyroglobulin levels in papillary thyroid carcinoma not prevent recurrences in patients with papillary
ment of well differentiated intrathyroid malignancy. after prophylactic unilateral central neck dissection. thyroid microcarcinoma. Clin Endocrinol (Oxf ).
Surgery. 2012;151:571–579. Ann Surg Oncol. 2012;19:1257–1263. 2013;78:614–620.
124. Lang BH, Wong CK. Lobectomy is a more cost- 143. Randolph GW, Duh QY, Heller KS, LiVolsi VA, 160. Baudin E, Travagli JP, Ropers J, Mancusi F,
effective option than total thyroidectomy for 1 to Mandel SJ, Steward DL, et al. The prognostic Bruno-Bossio G, Caillou B, et al. Microcarcinoma
4 cm papillary thyroid carcinoma that do not possess significance of nodal metastases from papillary of the thyroid gland: the Gustave-Roussy Institute
clinically recognizable high-risk features. Ann Surg thyroid carcinoma can be stratified based on the experience. Cancer. 1998;83:553–559.
Oncol. 2016;23:3641–3652. size and number of metastatic lymph nodes, as well 161. Creach KM, Siegel BA, Nussenbaum B, Grigsby PW.
125. Kluijfhout WP, Pasternak JD, Lim J, Kwon JS, as the presence of extranodal extension. Thyroid. Radioactive iodine therapy decreases recurrence in
Vriens MR, Clark OH, et al. Frequency of high- 2012;22:1144–1152. thyroid papillary microcarcinoma. ISRN Endocrinol.
risk characteristics requiring total thyroidectomy for 144. Moreno MA, Edeiken-Monroe BS, Siegel ER, 2012;2012:816386.
1-4 cm well-differentiated thyroid cancer. Thyroid. Sherman SI, Clayman GL. In papillary thyroid 162. Brown AP, Chen J, Hitchcock YJ. The risk of second
2016;26:820–824. cancer, preoperative central neck ultrasound detects primary malignancies up to three decades after the
126. Pinchot SN, Kunnimalaiyaan M, Sippel RS, Chen only macroscopic surgical disease, but negative find- treatment of differentiated thyroid cancer. J Clin
H. Medullary thyroid carcinoma: targeted therapies ings predict excellent long-term regional control and Endocrinol Metab. 2008;93:504–515.
and future directions. J Oncol. 2009;31:1830. survival. Thyroid. 2012;22:347–355. 163. Iyer NG, Morris LG, Tuttle RM. Rising incidence
127. Shaha AR, Shah JP, Loree TR. Low-risk differentiated 145. Gyorki DE, Untch B, Tuttle RM, Shaha AR. of second cancers in patients with low-risk (T1N0)
thyroid cancer: the need for selective treatment. Ann Prophylactic central neck dissection in differentiated thyroid cancer who receive radioactive iodine therapy.
Surg Oncol. 1997;4:328–333. thyroid cancer: an assessment of the evidence. Ann Cancer. 2011;117:4439–4446.
128. Wanebo H, Coburn M, Teates D, Cole B. Total Surg Oncol. 2013;20:2285–2289. 164. Chen AY, Levy L, Goepfert H. The development of
thyroidectomy does not enhance disease control or 146. Carty SE, Cooper DS, Doherty GM, Duh QY, breast carcinoma in women with thyroid carcinoma.
survival even in high-risk patients with differentiated et al. Consensus statement on the terminology and Cancer. 2001;92:225–231.
thyroid cancer. Ann Surg. 1998;227:912–921. classification of central neck dissection for thyroid 165. Wiklund TA, Blomqvist CP, Raty J. Postirradiation
129. Sanders LE, Cady B. Differentiated thyroid cancer: cancer. Thyroid. 2009;19:1153–1158. sarcoma. Analysis of a nationwide cancer registry
reexamination of risk groups and outcome of treat- 147. Tuttle RM, Ball DW, Byrd D. Thyroid carcinoma. material. Cancer. 1991;68:524–531.
ment. Arch Surg. 1998;133:419–425. J Natl Compr Canc Netw. 2010;8:1228–1274. 166. Mandel SJ, Mandel L. Radioactive iodine and the
130. Nguyen K, Dilawari RA. Predictive value of AMES 148. Reiners C, Dietlein M, Luster M. Radio-iodine salivary glands. Thyroid. 2003;13:265–271.
scoring system in selection of extent of surgery in therapy in differentiated thyroid cancer: indications 167. Almeida JP, Sanabria AE, Lima EN, Kowalski LP.
well differentiated carcinoma of thyroid. Am Surg. and procedures. Best Pract Res Clin Endocrinol Metab. Late side effects of radioactive iodine on salivary
1995;61:151–155. 2008;22:989–1007. gland function in patients with thyroid cancer. Head
131. Mazzaferri EL, Young RL. Papillary thyroid car- 149. Mazzaferri EL, Kloos RT. Using recombinant human Neck. 2011;33:686–690.
cinoma: a 10 year follow-up report of the impact TSH in the management of well-differentiated 168. Chakravarty D, Santos E, Ryder M, Knauf JA,
of therapy in 576 patients. Am J Med. 1981;70: thyroid cancer: current strategies and future direc- Liao XH, West BL, et al. Small-molecule MAPK
511–518. tions. Thyroid. 2000;10:767–778. inhibitors restore radioiodine incorporation in mouse
132. Samaan NA, Schultz PN, Hickey RC. The results of 150. Sawka AM, Thephamongkhol K, Brouwers M, thyroid cancers with conditional BRAF activation.
various modalities of treatment of well differentiated Thabane L, Browman G, Gerstein HC. Clinical J Clin Invest. 2011;121:4700–4711.
thyroid carcinomas: a retrospective review of 1599 review 170: a systematic review and metaanalysis 169. Elisei R, Viola D, Torregrossa L, Giannini R, Romei C,
patients. J Clin Endocrinol Metab. 1992;75:714– of the effectiveness of radioactive iodine remnant Ugolini C, et al. The BRAF(V600E) mutation is an
720. ablation for well-differentiated thyroid cancer. J Clin independent, poor prognostic factor for the outcome
133. Loh KC, Greenspan FS, Gee L. Pathological Endocrinol Metab. 2004;89:3668–3676. of patients with low-risk intrathyroid papillary
tumor-node-metastasis (pTNM) staging for papillary 151. Ma C, Xie J, Liu W. Recombinant human thy- thyroid carcinoma: single-institution results from a
and follicular thyroid carcinomas: a retrospective rotropin (rhTSH) aided radioiodine treatment for large cohort study. J Clin Endocrinol Metab. 2012;97:
analysis of 700 patients. J Clin Endocrinol Metab. residual or metastatic differentiated thyroid cancer. 4390–4398.
1997;82:3553–3562. Cochrane Database Syst Rev. 2010;(11):CD008302. 170. McGriff NJ, Csako G, Gourgiotis L. Effects of
134. Mazzaferri EL, Jhiang SM. Long-term impact of 152. Klubo-Gwiezdzinska J, Burman KD, Nostrand D. thyroid hormone suppression therapy on adverse
initial surgical and medical therapy on papillary Van Radioiodine treatment of metastatic thyroid clinical outcomes in thyroid cancer. Ann Med.
and follicular thyroid cancer. Am J Med. 1994;97: cancer: relative efficacy and side effect profile of 2002;34(7–8):554–564.
418–428. preparation by thyroid hormone withdrawal versus 171. Brierley JD, Tsang RW. External-beam radiation
135. Shindo M, Wu JC, Park EE, Tanzella F. The impor- recombinant human thyrotropin. Thyroid. 2012;22: therapy in the treatment of differentiated thyroid
tance of central compartment elective lymph node 310–317. cancer. Semin Surg Oncol. 1999;16:42–49.
excision in the staging and treatment of papillary 153. Jonklaas J, Sarlis NJ, Litofsky D. Outcomes of 172. Tennvall J, Lundell G, Hallquist A. Combined
thyroid cancer. Arch Otolaryngol Head Neck Surg. patients with differentiated thyroid carcinoma fol- doxorubicin, hyperfractionated radiotherapy, and
2006;132:650–654. lowing initial therapy. Thyroid. 2006;16:1229–1242. surgery in anaplastic thyroid carcinoma. Report
1107.e4 Part III: Specific Malignancies

on two protocols The Swedish Anaplastic Thyroid therapy. Surgery. 2013;154:1448–1454, discussion cytology in medullary thyroid carcinoma: a meta-
Cancer Group. Cancer. 1994;74:1348–1354. 54–55. analysis. Clin Endocrinol (Oxf ). 2015;82:280–285.
173. Shimaoka K, Schoenfeld DA, DeWys WD. A 189. Papini E, Bizzarri G, Bianchini A, Valle D, Misischi 207. Boi F, Maurelli I, Pinna G. Calcitonin measurement
randomized trial of doxorubicin versus doxorubicin I, Guglielmi R, et al. Percutaneous ultrasound-guided in wash-out fluid from fine needle aspiration of
plus cisplatin in patients with advanced thyroid laser ablation is effective for treating selected nodal neck masses in patients with primary and metastatic
carcinoma. Cancer. 1985;56:2155–2160. metastases in papillary thyroid cancer. J Clin medullary thyroid carcinoma. J Clin Endocrinol
174. Williams SD, Birch R, Einhorn LH. Phase, II. Endocrinol Metab. 2013;98:E92–E97. Metab. 2007;92:2115–2118.
evaluation of doxorubicin plus cisplatin in advanced 190. Wells SA Jr, Asa SL, Dralle H, Elisei R, Evans DB, 208. Chin WW, Goodman RH, Jacobs JW. Medullary
thyroid cancer: a Southeastern Cancer Study Group Gagel RF, et al. Revised American Thyroid Associa- thyroid carcinoma identified by cell-free translation
Trial. Cancer Treat Rep. 1986;70:405–407. tion guidelines for the management of medullary of tumor messenger ribonucleic acid in a patient
175. Higgins MJ, Forastiere A, Marur S. New directions in thyroid carcinoma. Thyroid. 2015;25:567–610. with a neck mass and the syndrome of ectopic
the systemic treatment of metastatic thyroid cancer. 191. Hill CS Jr, Ibanez ML, Samaan NA. Medullary adrenocorticotropin. J Clin Endocrinol Metab.
Oncology (Williston Park). 2009;23:768–775. (solid) carcinoma of the thyroid gland: an analysis 1981;52:572–575.
176. Kim DW, Jo YS, Jung HS. An orally administered of the M. D Anderson hospital experience with 209. Costante G, Meringolo D, Durante C. J. Predictive
multitarget tyrosine kinase inhibitor, SU11248, is patients with the tumor its special features and its value of serum calcitonin levels for preoperative
a novel potent inhibitor of thyroid oncogenic RET/ histogenesis. Medicine (Baltimore). 1973;52:141– diagnosis of medullary thyroid carcinoma in a cohort
papillary thyroid cancer kinases. J Clin Endocrinol 171. of 5817 consecutive patients with thyroid nodules.
Metab. 2006;91:4070–4076. 192. Roman S, Lin R, Sosa JA. Prognosis of medullary J Clin Endocrinol Metab. 2007;92:450–455.
177. Haugen BR, Kane MA. Approach to the thyroid thyroid carcinoma: demographic, clinical, and 210. Machens A, Schneyer U, Holzhausen HJ, Dralle
cancer patient with extracervical metastases. J Clin pathologic predictors of survival in 1252 cases. H. Prospects of remission in medullary thyroid
Endocrinol Metab. 2010;95:987–993. Cancer. 2006;107:2134–2142. carcinoma according to basal calcitonin level.
178. Brose MS, Nutting CM, Jarzab B, Elisei R, Siena 193. Kouvaraki MA, Shapiro SE, Fornage BD, Edeiken- J Clin Endocrinol Metab. 2005;90:2029–2034.
S, Bastholt L, et al. Sorafenib in radioactive iodine- Monro BS, Sherman SI, Vassilopoulou-Sellin R, 211. Machens A, Dralle H. Pretargeted anti-
refractory, locally advanced or metastatic differenti- et al. Role of preoperative ultrasonography in the carcinoembryonic-antigen radioimmunotherapy
ated thyroid cancer: a randomised, double-blind, surgical management of patients with thyroid cancer. for medullary thyroid carcinoma. J Clin Oncol.
phase 3 trial. Lancet. 2014;384:319–328. Surgery. 2003;134:946–954, discussion 54–55. 2006;24:e37, author reply e38.
179. Schlumberger M, Tahara M, Wirth LJ, Robinson B, 194. Wells SA Jr, Pacini F, Robinson BG, Santoro M. 212. de Groot JW, Kema IP, Breukelman H. Biochemical
Brose MS, Elisei R, et al. Lenvatinib versus placebo Multiple endocrine neoplasia type 2 and familial markers in the follow-up of medullary thyroid cancer.
in radioiodine-refractory thyroid cancer. N Engl J medullary thyroid carcinoma: an update. J Clin Thyroid. 2006;16:1163–1170.
Med. 2015;372:621–630. Endocrinol Metab. 2013;98:3149–3164. 213. Wells SA Jr, Chi DD, Toshima K. Predictive DNA.
180. Tuttle RM, Tala H, Shah J. Estimating risk of 195. Roy M, Chen H, Sippel RS. Current understand- Wel testing and prophylactic thyroidectomy in
recurrence in differentiated thyroid cancer after ing and management of medullary thyroid cancer. patients at risk for multiple endocrine neoplasia
total thyroidectomy and radioactive iodine remnant Oncologist. 2013;18:1093–1100. type 2A. Ann Surg. 1994;220:237–247, discussion
ablation: using response to therapy variables to 196. Machens A, Hauptmann S, Dralle H. Increased risk 247250.
modify the initial risk estimates predicted by the of lymph node metastasis in multifocal hereditary 214. Ciampi R, Mian C, Fugazzola L, Cosci B, Romei
new American Thyroid Association staging system. and sporadic medullary thyroid cancer. World J Surg. C, Barollo S, et al. Evidence of a low prevalence of
Thyroid. 2010;20:1341–1349. 2007;31:1960–1965. RAS mutations in a large medullary thyroid cancer
181. Hay ID. Management of patients with low-risk papil- 197. Kaserer K, Scheuba C, Neuhold N, Weinhausel series. Thyroid. 2013;23:50–57.
lary thyroid carcinoma. Endocr Pract. 2007;13:521– A, Haas OA, Vierhapper H, et al. Sporadic versus 215. Moura MM, Cavaco BM, Pinto AE, Leite V. High
533. familial medullary thyroid microcarcinoma: a prevalence of RAS mutations in RET-negative
182. Lesnik D, Cunnane ME, Zurakowski D, Acar histopathologic study of 50 consecutive patients. sporadic medullary thyroid carcinomas. J Clin
GO, Ecevit C, Mace A, et al. Papillary thyroid Am J Surg Pathol. 2001;25:1245–1251. Endocrinol Metab. 2011;96:E863–E868.
carcinoma nodal surgery directed by a preoperative 198. Miyauchi A, Matsuzuka F, Hirai K, Yokozawa 216. Boichard A, Croux L, Al Ghuzlan A, Broutin
radiographic map utilizing CT scan and ultrasound T, Kobayashi K, Ito Y, et al. Prospective trial of S, Dupuy C, Leboulleux S, et al. Somatic RAS
in all primary and reoperative patients. Head Neck. unilateral surgery for nonhereditary medullary mutations occur in a large proportion of sporadic
2014;36:191–202. thyroid carcinoma in patients without germline RET-negative medullary thyroid carcinomas and
183. Yeh MW, Bauer AJ, Bernet VA, Ferris RL, Loevner RET mutations. World J Surg. 2002;26:1023–1028. extend to a previously unidentified exon. J Clin
LA, Mandel SJ, et al. American Thyroid Association 199. Sippel RS, Kunnimalaiyaan M, Chen H. Current Endocrinol Metab. 2012;97:E2031–E2035.
statement on preoperative imaging for thyroid cancer management of medullary thyroid cancer. Oncologist. 217. Moura MM, Cavaco BM, Pinto AE, Domingues
surgery. Thyroid. 2015;25:3–14. 2008;13:539–547. R, Santos JR, Cid MO, et al. Correlation of RET
184. Tufano RP, Clayman G, Heller KS, Inabnet WB, 200. Chen H, Sippel RS, O’Dorisio MS, Vinik AI, Lloyd somatic mutations with clinicopathological features
Kebebew E, Shaha A, et al. Management of RV, Pacak K. The North American Neuroendocrine in sporadic medullary thyroid carcinomas. Br J
recurrent/persistent nodal disease in patients with Tumor Society consensus guideline for the diag- Cancer. 2009;100:1777–1783.
differentiated thyroid cancer: a critical review of nosis and management of neuroendocrine tumors: 218. Elisei R, Cosci B, Romei C, Bottici V, Renzini G,
the risks and benefits of surgical intervention versus pheochromocytoma, paraganglioma, and medullary Molinaro E, et al. Prognostic significance of somatic
active surveillance. Thyroid. 2015;25:15–27. thyroid cancer. Pancreas. 2010;39:775–783. RET oncogene mutations in sporadic medullary
185. Phelan E, Kamani D, Shin J, Randolph GW. Neural 201. Williams ED, Brown CL, Doniach I. Pathologi- thyroid cancer: a 10-year follow-up study. J Clin
monitored revision thyroid cancer surgery: surgical cal and clinical findings in a series of 67 cases of Endocrinol Metab. 2008;93:682–687.
safety and thyroglobulin response. Otolaryngol Head medullary carcinoma of the thyroid; 1966. 219. Kouvaraki MA, Shapiro SE, Perrier ND. RET
Neck Surg. 2013;149:47–52. 202. Szavcsur P, Godeny M, Bajzik G, Lengyel E, Repa proto-oncogene: a review and update of genotype-
186. Urken ML, Milas M, Randolph GW, Tufano I, Tron L, et al. Angiography-proven liver metastases phenotype correlations in hereditary medullary
R, Bergman D, Bernet V, et al. Management of explain low efficacy of lymph node dissections in thyroid cancer and associated endocrine tumors.
recurrent and persistent metastatic lymph nodes in medullary thyroid cancer patients. Eur J Surg Oncol. Thyroid. 2005;15:531–544.
well-differentiated thyroid cancer: a multifactorial 2005;31:183–190. 220. Ogilvie JB, Kebebew E. Indication and timing of
decision-making guide for the Thyroid Cancer Care 203. Giraudet AL, Vanel D, Leboulleux S, Auperin A, thyroid surgery for patients with hereditary medul-
Collaborative. Head Neck. 2015;37:605–614. Dromain C, Chami L, et al. Imaging medullary lary thyroid cancer syndromes. J Natl Compr Canc
187. Heilo A, Sigstad E, Fagerlid KH, Haskjold OI, thyroid carcinoma with persistent elevated calcitonin Netw. 2006;4:139–147.
Groholt KK, Berner A, et al. Efficacy of ultrasound- levels. J Clin Endocrinol Metab. 2007;92:4185–4190. 221. Moley JF, DeBenedetti MK. Patterns of nodal
guided percutaneous ethanol injection treatment in 204. Bugalho MJ, Santos JR, Sobrinho L. Preoperative metastases in palpable medullary thyroid carcinoma:
patients with a limited number of metastatic cervical diagnosis of medullary thyroid carcinoma: fine needle recommendations for extent of node dissection. Ann
lymph nodes from papillary thyroid carcinoma. aspiration cytology as compared with serum calci- Surg. 1999;229:880–887, discussion 887888.
J Clin Endocrinol Metab. 2011;96:2750– tonin measurement. J Surg Oncol. 2005;91:56–60. 222. Greenblatt DY, Elson D, Mack E, Chen H. Initial
2755. 205. Bhanot P, Yang J, Schnadig VJ, Logrono R. Role lymph node dissection increases cure rates in
188. Hay ID, Lee RA, Davidge-Pitts C, Reading CC, of FNA cytology and immunochemistry in the patients with medullary thyroid cancer. Asian J
Charboneau JW. Long-term outcome of ultrasound- diagnosis and management of medullary thyroid Surg. 2007;30:108–112.
guided percutaneous ethanol ablation of selected carcinoma: report of six cases and review of the 223. Machens A, Holzhausen HJ, Dralle H. Contralateral
“recurrent” neck nodal metastases in 25 patients literature. Diagn Cytopathol. 2007;35:285–292. cervical and mediastinal lymph node metastasis in
with TNM stages III or IVA papillary thyroid 206. Trimboli P, Treglia G, Guidobaldi L, Romanelli F, medullary thyroid cancer: systemic disease? Surgery.
carcinoma previously treated by surgery and 131I Nigri G, Valabrega S, et al. Detection rate of FNA 2006;139:28–32.
Cancer of the Endocrine System  •  CHAPTER 68 1107.e5
1107.e5

224. Machens A, Dralle H. Biomarker-based risk strati- 244. Barreau O, Assie G, Wilmot-Roussel H, Ragazzon cancer (ACC)—literature overview and own experi-
fication for previously untreated medullary thyroid B, Baudry C, Perlemoine K, et al. Identification ence. Endokrynol Pol. 2014;65:492–502.
cancer. J Clin Endocrinol Metab. 2010;95:2655–2663. of a CpG island methylator phenotype in adre- 266. Brix D, Allolio B, Fenske W. Laparoscopic versus
225. Machens A, Dralle H. Genotype-phenotype based nocortical carcinomas. J Clin Endocrinol Metab. open adrenalectomy for adrenocortical carcinoma:
surgical concept of hereditary medullary thyroid 2013;98:E174–E184. surgical and oncologic outcome in 152 patients.
carcinoma. World J Surg. 2007;31:957–968. 245. Libe R, Fratticci A, Bertherat J. Adrenocortical Eur Urol. 2010;58:609–615.
226. Tischler AS, Khan A, DeLellis RA. Prophylactic cancer: pathophysiology and clinical management. 267. Schteingart DE, Doherty GM, Gauger PG, Giordano
thyroidectomy in multiple endocrine neoplasia type Endocr Relat Cancer. 2007;14:13–28. TJ, Hammer GD, Korobkin M, et al. Management
2A. N Engl J Med. 2005;353:2817–2818, author 246. Kjellman M, Roshani L, Teh BT. Genotyping of of patients with adrenal cancer: recommendations
reply 2817–2818. adrenocortical tumors: very frequent deletions of the of an international consensus conference. Endocr
227. Chen H, Roberts JR, Ball DW. Effective long-term MEN1 locus in 11q13 and of a 1-centimorgan region Relat Cancer. 2005;12:667–680.
palliation of symptomatic, incurable metastatic in 2p16. J Clin Endocrinol Metab. 1999;84:730–735. 268. Reibetanz J, Jurowich C, Erdogan I, Nies C, Rayes N,
medullary thyroid cancer by operative resection. 247. Bertherat J, Bertagna X. Pathogenesis of adrenocorti- Dralle H, et al. Impact of lymphadenectomy on the
Ann Surg. 1998;227:887–895. cal cancer. Best Pract Res Clin Endocrinol Metab. oncologic outcome of patients with adrenocortical
228. Barbet J, Campion L, Kraeber-Bodere F, Chatal 2009;23:261–271. carcinoma. Ann Surg. 2012;255:363–369.
JF. Prognostic impact of serum calcitonin and 248. Wandoloski M, Bussey KJ, Demeure MJ. Adre- 269. Sbiera S, Leich E, Liebisch G, Sbiera I, Schirbel A,
carcinoembryonic antigen doubling-times in patients nocortical cancer. Surg Clin North Am. 2009;89: Wiemer L, et al. Mitotane inhibits sterol-O-acyl
with medullary thyroid carcinoma. J Clin Endocrinol 1255–1267. transferase 1 triggering lipid-mediated endoplasmic
Metab. 2005;90:6077–6084. 249. Fassnacht M, Libe R, Kroiss M, Allolio B. Adre- reticulum stress and apoptosis in adrenocortical car-
229. Ball DW. Medullary thyroid cancer: therapeutic nocortical carcinoma: a clinician’s update. Nat Rev cinoma cells. Endocrinology. 2015;156:3895–3908.
targets and molecular markers. Curr Opin Oncol. Endocrinol. 2011;7:323–335. 270. Terzolo M, Angeli A, Fassnacht M. Adjuvant
2007;19:18–23. 250. Czajka I, Zgliczynski W, Kasperlik-Zaluska A, mitotane treatment for adrenocortical carcinoma.
230. Wells SA Jr, Robinson BG, Gagel RF. Wel Vandetanib Cichocki A. [Gynecomasty as a first sign of adrenal N Engl J Med. 2007;356:2372–2380.
in patients with locally advanced or metastatic med- carcinoma—case report]. Endokrynol Pol. 2005;56: 271. Grubbs EG, Callender GG, Xing Y. Recurrence
ullary thyroid cancer: a randomized, double-blind 940–944. of adrenal cortical carcinoma following resection:
phase III trial. J Clin Oncol. 2012;30:134–141. 251. Stewart JN, Flageole H, Kavan P. A surgical approach surgery alone can achieve results equal to surgery
231. de Groot JW, Zonnenberg BA, van Ufford-Mannesse to adrenocortical tumors in children: the mainstay plus mitotane. Ann Surg Oncol. 2010;17:263–270.
PQ. A phase II trial of imatinib therapy for meta- of treatment. J Pediatr Surg. 2004;39:759–763. 272. Baudin E, Leboulleux S, Al Ghuzlan A. Therapeutic
static medullary thyroid carcinoma. J Clin Endocrinol 252. Fassnacht M, Allolio B. Clinical management management of advanced adrenocortical carcinoma:
Metab. 2007;92:3466–3469. of adrenocortical carcinoma. Best Pract Res Clin what do we know in 2011? Horm Cancer. 2011;2:
232. Langmuir PB, Yver A. Vandetanib for the treat- Endocrinol Metab. 2009;23:273–289. 363–371.
ment of thyroid cancer. Clin Pharmacol Ther. 253. Young WF Jr. Clinical practice. The inciden- 273. Terzolo M, Berruti A. Adjunctive treatment of
2012;91:71–80. tally discovered adrenal mass. N Engl J Med. adrenocortical carcinoma. Curr Opin Endocrinol
233. Kurzrock R, Sherman SI, Ball DW, Forastiere AA, 2007;356:601–610. Diabetes Obes. 2008;15:221–226.
Cohen RB, Mehra R, et al. Activity of XL184 254. Copeland PM. The incidentally discovered adrenal 274. Hahner S, Fassnacht M. Mitotane for adrenocortical
(Cabozantinib), an oral tyrosine kinase inhibitor, mass. Ann Surg. 1984;199:116–122. carcinoma treatment. Curr Opin Investig Drugs.
in patients with medullary thyroid cancer. J Clin 255. Grumbach MM, Biller BM, Braunstein GD. 2005;6:386–394.
Oncol. 2011;29:2660–2666. Management of the clinically inapparent adrenal 275. Fassnacht M, Terzolo M, Allolio B. Combination
234. Soon PS, Sidhu SB. Adrenocortical carcinoma. mass (“incidentaloma”). Ann Intern Med. 2003;138: chemotherapy in advanced adrenocortical carcinoma.
Cancer Treat Res. 2010;153:187–210. 424–429. N Engl J Med. 2012;366:2189–2197.
235. Lafemina J, Brennan MF. Adrenocortical carcinoma: 256. Petersenn S, Richter PA, Broemel T, Ritter 276. Fassnacht M, Hahner S, Polat B. Efficacy of adjuvant
past, present, and future. J Surg Oncol. 2012;106: CO, Deutschbein T, Beil FU, et al. Computed radiotherapy of the tumor bed on local recurrence of
586–594. tomography criteria for discrimination of adrenal adrenocortical carcinoma. J Clin Endocrinol Metab.
236. Bilimoria KY, Shen WT, Elaraj D. Adrenocortical adenomas and adrenocortical carcinomas: analysis 2006;91:4501–4504.
carcinoma in the United States: treatment utilization of the German ACC registry. Eur J Endocrinol. 277. Ho J, Turkbey B, Edgerly M, Alimchandani
and prognostic factors. Cancer. 2008;113:3130– 2015;172:415–422. M, Quezado M, Camphausen K, et al. Role of
3136. 257. Pena CS, Boland GW, Hahn PF. Characterization radiotherapy in adrenocortical carcinoma. Cancer
237. Icard P, Goudet P, Charpenay C. Adrenocortical car- of indeterminate (lipid-poor) adrenal masses: use J. 2013;19:288–294.
cinomas: surgical trends and results of a 253-patient of washout characteristics at contrast-enhanced CT. 278. Habra MA, Ejaz S, Feng L, Das P, Deniz F, Grubbs
series from the French Association of Endocrine Radiology. 2000;217:798–802. EG, et al. A retrospective cohort analysis of the effi-
Surgeons study group. World J Surg. 2001;25: 258. Young WF Jr. Conventional imaging in adrenocorti- cacy of adjuvant radiotherapy after primary surgical
891–897. cal carcinoma: update and perspectives. Horm Cancer. resection in patients with adrenocortical carcinoma.
238. Stojadinovic A, Ghossein RA, Hoos A. Adrenocortical 2011;2:341–347. J Clin Endocrinol Metab. 2013;98:192–197.
carcinoma: clinical, morphologic, and molecular 259. Boland GW, Dwamena BA, Jagtiani Sangwaiya M, 279. Hermsen IG, Groenen YE, Dercksen MW, Theuws
characterization. J Clin Oncol. 2002;20:941–950. Goehler AG, Blake MA, Hahn PF, et al. Charac- J, Haak HR. Response to radiation therapy in adre-
239. Michalkiewicz E, Sandrini R, Figueiredo B. Clinical terization of adrenal masses by using FDG PET: a nocortical carcinoma. J Endocrinol Invest. 2010;33:
and outcome characteristics of children with adre- systematic review and meta-analysis of diagnostic 712–714.
nocortical tumors: a report from the International test performance. Radiology. 2011;259:117–126. 280. Sabolch A, Feng M, Griffith K, Hammer G, Doherty
Pediatric Adrenocortical Tumor Registry. J Clin 260. Gust L, Taieb D, Beliard A, Barlier A, Morange I, G, Ben-Josef E. Adjuvant and definitive radiotherapy
Oncol. 2004;22:838–845. de Micco C, et al. Preoperative 18F-FDG uptake for adrenocortical carcinoma. Int J Radiat Oncol Biol
240. Latronico AC, Pinto EM, Domenice S, Fragoso is strongly correlated with malignancy, Weiss score, Phys. 2011;80:1477–1484.
MC, Martin RM, Zerbini MC, et al. An inherited and molecular markers of aggressiveness in adrenal 281. Polat B, Fassnacht M, Pfreundner L. Radiotherapy
mutation outside the highly conserved DNA-binding cortical tumors. World J Surg. 2012;36:1406– in adrenocortical carcinoma. Cancer. 2009;115:
domain of the p53 tumor suppressor protein in chil- 1410. 2816–2823.
dren and adults with sporadic adrenocortical tumors. 261. Hahner S, Sundin A. Metomidate-based imaging 282. Allolio B, Fassnacht M. Clinical review: adrenocorti-
J Clin Endocrinol Metab. 2001;86:4970–4973. of adrenal masses. Horm Cancer. 2011;2:348–353. cal carcinoma: clinical update. J Clin Endocrinol
241. Zhang J, Walsh MF, Wu G, Edmonson MN, 262. Hahner S, Kreissl MC, Fassnacht M, Haenscheid H, Metab. 2006;91:2027–2037.
Gruber TA, Easton J, et al. Germline mutations Bock S, Verburg FA, et al. Functional characteriza- 283. Fassnacht M, Berruti A, Baudin E, Demeure MJ,
in predisposition genes in pediatric cancer. N Engl tion of adrenal lesions using [123I]IMTO-SPECT/ Gilbert J, Haak H, et al. Linsitinib (OSI-906)
J Med. 2015;373:2336–2346. CT. J Clin Endocrinol Metab. 2013;98:1508–1518. versus placebo for patients with locally advanced
242. Gaujoux S, Grabar S, Fassnacht M, Ragazzon B, 263. Kreissl MC, Schirbel A, Fassnacht M, Haenscheid or metastatic adrenocortical carcinoma: a double-
Launay P, Libe R, et al. beta-catenin activation H, Verburg FA, Bock S, et al. [(1)(2)(3)I]Iodome- blind, randomised, phase 3 study. Lancet Oncol.
is associated with specific clinical and pathologic tomidate imaging in adrenocortical carcinoma. 2015;16:426–435.
characteristics and a poor outcome in adrenocortical J Clin Endocrinol Metab. 2013;98:2755–2764. 284. Schulick RD, Brennan MF. Long-term survival after
carcinoma. Clin Cancer Res. 2011;17:328–336. 264. Balasubramaniam S, Fojo T. Practical considerations complete resection and repeat resection in patients
243. Assie G, Letouze E, Fassnacht M, Jouinot A, in the evaluation and management of adrenocortical with adrenocortical carcinoma. Ann Surg Oncol.
Luscap W, Barreau O, et al. Integrated genomic cancer. Semin Oncol. 2010;37:619–626. 1999;6:719–726.
characterization of adrenocortical carcinoma. Nat 265. Dworakowska D, Drabarek A, Wenzel I, Babinska A, 285. Fassnacht M, Johanssen S, Fenske W, Weismann
Genet. 2014;46:607–612. Swiatkowska-Stodulska R, Sworczak K. Adrenocortical D, Agha A, Beuschlein F, et al. Improved survival
1107.e6 Part III: Specific Malignancies

in patients with stage II adrenocortical carcinoma 307. Strong VE, Kennedy T, Al-Ahmadie H. Prognostic 325. Wermer P. Genetic aspects of adenomatosis of
followed up prospectively by specialized centers. indicators of malignancy in adrenal pheochromocyto- endocrine glands. Am J Med. 1954;16:363–371.
J Clin Endocrinol Metab. 2010;95:4925–4932. mas: clinical, histopathologic, and cell cycle/apoptosis 326. Zollinger RM, Ellison EH. Primary peptic ulcerations
286. Harari A, Inabnet WB 3rd. Malignant pheochro- gene expression analysis. Surgery. 2008;143:759–768. of the jejunum associated with islet cell tumors of the
mocytoma a review. Am J Surg. 2011;201:700–708. 308. Thompson LD. Pheochromocytoma of the Adrenal pancreas. Ann Surg. 1955;142:709–723, discussion
287. Grogan RH, Mitmaker EJ, Duh QY. Changing gland Scaled Score (PASS) to separate benign from 724–728.
paradigms in the treatment of malignant pheo- malignant neoplasms: a clinicopathologic and 327. Lulu DJ, Corcoran TE, Andre M. Familial
chromocytoma. Cancer Control. 2011;18:104– immunophenotypic study of 100 cases. Am J Clin endocrine adenomatosis with associated Zollinger-
112. Pathol. 2002;26:551–566. Ellison syndrome. Wermers syndrome. Am J Surg.
288. Zarnegar R, Kebebew E, Duh QY, Clark OH. 309. Zografos GN, Farfaras A, Vasiliadis G. Laparoscopic 1968;115:695–701.
Malignant pheochromocytoma. Surg Oncol Clin. resection of large adrenal tumors. JSLS. 2010;14: 328. Marx SJ. Multiple endocrine neoplasia type 12002.
2006;15:555–571. 364–368. 329. Thakker RV, Newey PJ, Walls GV, Bilezikian J, Dralle
289. Adjalle R, Plouin PF, Pacak K, Lehnert H. Treatment 310. Zografos GN, Vasiliadis G, Farfaras AN. Laparo- H, Ebeling PR, et al. Clinical practice guidelines
of malignant pheochromocytoma. Horm Metab Res. scopic surgery for malignant adrenal tumors. JSLS. for multiple endocrine neoplasia type 1 (MEN1).
2009;41:687–696. 2009;13:196–202. J Clin Endocrinol Metab. 2012;97:2990–3011.
290. Kantorovich V, King KS, Pacak K. SDH-related 311. Adler JT, Chen H, Schaefer S, Sippel RS. What is 330. Schussheim DH, Skarulis MC, Agarwal SK. Multiple
pheochromocytoma and paraganglioma. Best Pract the added benefit of cervical ultrasound to (99m) endocrine neoplasia type 1: new clinical and basic
Res Clin Endocrinol Metab. 2010;24:415–424. Tc-sestamibi scanning in primary hyperparathyroid- findings. Trends Endocrinol Metab. 2001;12:173–178.
291. Neumann HP, Bausch B, McWhinney SR. Germ-line ism? Ann Surg Oncol. 2011;18:2907–2911. 331. Pipeleers-Marichal M, Somers G, Willems G, Foulis
mutations in nonsyndromic pheochromocytoma. 312. Kinney MA, Warner ME, vanHeerden JA. A, Imrie C, Bishop AE, et al. Gastrinomas in the
N Engl J Med. 2002;346:1459–1466. Perianesthetic risks and outcomes of pheochromo- duodenums of patients with multiple endocrine
292. Neumann HP, Pawlu C, Peczkowska M. Distinct cytoma and paraganglioma resection. Anesth Analg. neoplasia type 1 and the Zollinger-Ellison syndrome.
clinical features of paraganglioma syndromes associ- 2000;91:1118–1123. N Engl J Med. 1990;322:723–727.
ated with SDHB and SDHD gene mutations. JAMA. 313. Mukherjee JJ, Kaltsas GA, Islam N, Plowman PN, 332. Akerstrom G, Stalberg P. Surgical management of
2004;292:943–951. Foley R, Hikmat J, et al. Treatment of metastatic MEN-1 and- state of the art. Surg Clin North Am.
293. DeLellis RA, Lloyd RV, Heitz PU, Eng C (eds). carcinoid tumours, phaeochromocytoma, paragan- 2009;89(2):1047–1068.
Pathology and Genetics of Tumours of Endocrine glioma and medullary carcinoma of the thyroid with 333. Gibril F, Schumann M, Pace A, Jensen RT. Multiple
Organs. Lyon, France: IARC press; 2004. (131)I-meta-iodobenzylguanidine [(131)I-mIBG]. endocrine neoplasia type 1 and Zollinger-Ellison
294. Adler JT, Meyer-Rochow GY, Chen H. Pheo- Clin Endocrinol (Oxf ). 2001;55:47–60. syndrome: a prospective study of 107 cases and
chromocytoma: current approaches and future 314. Rutherford MA, Rankin AJ, Yates TM, Mark PB, comparison with 1009 cases from the literature.
directions. Oncologist. 2008;13:779–793. Perry CG, Reed NS, et al. Management of metastatic Medicine (Baltimore). 2004;83:43–83.
295. Baguet JP, Hammer L, Mazzuco TL. Circumstances phaeochromocytoma and paraganglioma: use of 334. Rubinstein WS. Endocrine cancer predisposition
of discovery of phaeochromocytoma: a retrospective iodine-131-meta-iodobenzylguanidine therapy in syndromes: hereditary paraganglioma, multiple
study of 41 consecutive patients. Eur J Endocrinol. a tertiary referral centre. QJM. 2015;108:361–368. endocrine neoplasia type 1, multiple endocrine
2004;150:681–686. 315. Huang H, Abraham J, Hung E. Treatment of neoplasia type 2, and hereditary thyroid cancer.
296. Beuschlein F. Adrenal incidentalomas: presentation malignant pheochromocytoma/paraganglioma with Hematol Oncol Clin North Am. 2010;24:907–
and clinical work-up. Horm Res. 2007;68(suppl 5): cyclophosphamide, vincristine, and dacarbazine: 937.
191–194. recommendation from a 22-year follow-up of 18 335. Brandi ML, Gagel RF, Angeli A. Guidelines for
297. Tischler AS. Pheochromocytoma and extra-adrenal patients. Cancer. 2008;113:2020–2028. diagnosis and therapy of MEN type 1 and type 2.
paraganglioma: updates. Arch Pathol Lab Med. 2008; 316. Niemeijer ND, Alblas G, van Hulsteijn LT, J Clin Endocrinol Metab. 2001;86:5658–5671.
132:1272–1284. Dekkers OM, Corssmit EP. Chemotherapy with 336. Akerstrom G, Johansson H, Grama D. Surgical treat-
298. Chrisoulidou A, Kaltsas G, Ilias I, Grossman AB. cyclophosphamide, vincristine and dacarbazine for ment of endocrine pancreatic lesions in MEN-1. Acta
The diagnosis and management of malignant malignant paraganglioma and pheochromocytoma: Oncol. 1991;30:541–545.
phaeochromocytoma and paraganglioma. Endocr systematic review and meta-analysis. Clin Endocrinol 337. Sheppard BC, Norton JA, Doppman JL, Maton PN,
Relat Cancer. 2007;14:569–585. (Oxf ). 2014;81:642–651. Gardner JD, Jensen RT. Management of islet cell
299. Prejbisz A, Lenders JW, Eisenhofer G, Januszewicz 317. Harari A, Inabnet WB 3rd. Malignant pheochro- tumors in patients with multiple endocrine neoplasia:
A. Cardiovascular manifestations of phaeochromo- mocytoma: a review. Am J Surg. 2011;201:700–708. a prospective study. Surgery. 1989;106:1108–1117,
cytoma. J Hypertens. 2011;29:2049–2060. 318. Forrer F, Riedweg I, Maecke HR, Mueller-Brand J. discussion 17–18.
300. Walther MM, Reiter R, Keiser HR. Clinical and Radiolabeled DOTATOC in patients with advanced 338. Doherty GM. Multiple endocrine neoplasia type
genetic characterization of pheochromocytoma in paraganglioma and pheochromocytoma. Q J Nucl 1. J Surg Oncol. 2005;89:143–150.
von Hippel-Lindau families: comparison with spo- Med Mol Imaging. 2008;52:334–340. 339. Thompson NW. Current concepts in the surgical
radic pheochromocytoma gives insight into natural 319. Reisch N, Peczkowska M, Januszewicz A, Neumann management of multiple endocrine neoplasia type 1
history of pheochromocytoma. J Urol. 1999;162: HP. Pheochromocytoma: presentation, diagnosis and pancreatic-duodenal disease. Results in the treatment
659–664. treatment. J Hypertens. 2006;24:2331-2339. of 40 patients with Zollinger-Ellison syndrome,
301. Pomares FJ, Canas R, Rodriguez JM. Differences 320. Ayala-Ramirez M, Chougnet CN, Habra MA, Palmer hypoglycaemia or both. J Intern Med. 1998;243:
between sporadic and multiple endocrine neoplasia JL, Leboulleux S, Cabanillas ME, et al. Treatment 495–500.
type 2A phaeochromocytoma. Clin Endocrinol (Oxf). with sunitinib for patients with progressive metastatic 340. Hausman MS Jr, Thompson NW, Gauger PG,
1998;48:195–200. pheochromocytomas and sympathetic paragan- Doherty GM. The surgical management of MEN-1
302. Guller U, Turek J, Eubanks S. Detecting pheochro- gliomas. J Clin Endocrinol Metab. 2012;97:4040– pancreatoduodenal neuroendocrine disease. Surgery.
mocytoma: defining the most sensitive test. Ann 4050. 2004;136:1205–1211.
Surg. 2006;243:102–107. 321. Szalat A, Fraenkel M, Doviner V, Salmon A, Gross 341. Demeure MJ, Klonoff DC, Karam JH, Duh QY,
303. Taylor RL, Singh RJ. Validation of liquid DJ. Malignant pheochromocytoma: predictive factors Clark OH. Insulinomas associated with multiple
chromatography-tandem mass spectrometry method of malignancy and clinical course in 16 patients at endocrine neoplasia type I: the need for a different
for analysis of urinary conjugated metanephrine and a single tertiary medical center. Endocrine. 2011;39: surgical approach. Surgery. 1991;110:998–1004,
normetanephrine for screening of pheochromocy- 160–166. discussion -5.
toma. Clin Chem. 2002;48:533–539. 322. Goffredo P, Sosa JA, Roman SA. Malignant pheo- 342. Schimke RN, Hartmann WH. Familial amyloid-
304. Chen H, Sippel RS, O’Dorisio MS. The North chromocytoma and paraganglioma: a population producing medullary thyroid carcinoma and
American Neuroendocrine Tumor Society consensus level analysis of long-term survival over two decades. pheochromocytoma. A distinct genetic entity. Ann
guideline for the diagnosis and management of neu- J Surg Oncol. 2013;107:659–664. Intern Med. 1965;63:1027–1039.
roendocrine tumors: pheochromocytoma, paragangli- 323. Andersen KF, Altaf R, Krarup-Hansen A. Malignant 343. Bergholm U, Adami HO, Telenius-Berg M. Incidence
oma, and medullary thyroid cancer. Pancreas. 2010; pheochromocytomas and paragangliomas—the of sporadic and familial medullary thyroid carcinoma
39:775–783. importance of a multidisciplinary approach. Cancer in Sweden through. A nationwide study in 126
305. Greenblatt DY, Shenker Y, Chen H. The utility of Treat Rev. 2011;37:111–119. patients Swedish Group. Acta Oncol. 1990;29:9–15.
metaiodobenzylguanidine (MIBG) scintigraphy in 324. Pacak K, Eisenhofer G, Ahlman H, Bornstein 344. Callender GG, Rich TA, Perrier ND. Multiple
patients with pheochromocytoma. Ann Surg Oncol. SR, Gimenez-Roqueplo AP, Grossman AB, et al. endocrine neoplasia syndromes. Surg Clin North
2008;15:900–905. Pheochromocytoma: recommendations for clinical Am. 2008;88:863–895, viii.
306. Ohta S, Lai EW, Pang AL. Downregulation of practice from the First International Symposium. 345. Vasen HF, van Raue F, der Feltz M. The natural course
metastasis suppressor genes in malignant pheo- October 2005. Nat Clin Pract Endocrinol Metab. of multiple endocrine neoplasia type IIb. A study of
chromocytoma. Int J Cancer. 2005;114:139–143. 2007;3:92–102. 18 cases. Arch Intern Med. 1992;152:1250–1252.
Cancer of the Endocrine System  •  CHAPTER 68 1107.e7
1107.e7

346. Thosani S, Ayala-Ramirez M, Palmer L, Hu MI, 366. Rindi G, Kloppel G, Alhman H. TNM staging 386. Thorson A, Biorck G, Bjorkman G, Waldenstrom J,
Rich T, Gagel RF, et al. The characterization of of foregut (neuro)endocrine tumors: a consensus Heart J. Malignant carcinoid of the small intestine
pheochromocytoma and its impact on overall proposal including a grading system. Virchows Arch. with metastases to the liver, valvular disease of the
survival in multiple endocrine neoplasia type 2. J 2006;449:395–401. right side of the heart (pulmonary stenosis and tricus-
Clin Endocrinol Metab. 2013;98:E1813–E1819. 367. Rindi G, Kloppel G, Couvelard A. TNM staging pid regurgitation without septal defects), peripheral
347. Webb TA, Sheps SG, Carney JA. Differences between of midgut and hindgut (neuro) endocrine tumors: vasomotor symptoms, bronchoconstriction, and an
sporadic pheochromocytoma and pheochromocy- a consensus proposal including a grading system. unusual type of cyanosis; a clinical and pathologic
toma in multiple endocrine neoplasia, type 2. Am Virchows Arch. 2007;451:757–762. syndrome. Am Heart J. 1954;47:795–817.
J Surg Pathol. 1980;4:121–126. 368. Pahlavan PS, Kanthan R. Goblet cell carcinoid of 387. Pernow B, Waldenstrom J. Paroxysmal flushing and
348. Machens A, Lorenz K, Dralle H. Peak incidence of the appendix. World J Surg Oncol. 2005;3:36. other symptoms caused by 5-hydroxytryptamine
pheochromocytoma and primary hyperparathyroid- 369. Gould VE, Linnoila RI, Memoli VA, Warren WH. and histamine in patients with malignant tumours.
ism in multiple endocrine neoplasia 2: need for Neuroendocrine components of the bronchopulmo- Lancet. 1954;951:267.
age-adjusted biochemical screening. J Clin Endocrinol nary tract: hyperplasias, dysplasias, and neoplasms. 388. Oates JA, Melmon K, Sjoerdsma A. Release of a
Metab. 2013;98:E336–E345. Lab Invest. 1983;49:519–537. kinin peptide in the carcinoid syndrome. Lancet.
349. Myers SM, Eng C, Ponder BA, Mulligan LM. 370. Schein P. C. B. editor. Decision Making in Oncology; 1964;1:514–517.
Characterization of RET proto-oncogene 3’ 1988. 389. Oates JA, Pettinger WA, Doctor RB. Evidence for
splicing variants and polyadenylation sites: a novel 371. Deacon AC. The measurement of 5-hydroxyindole- the release of bradykinin in carcinoid syndrome.
C-terminus for RET. Oncogene. 1995;11:2039– acetic acid in urine. Ann Clin Biochem. 1994;31(Pt J Clin Invest. 1966;45:173–178.
2045. 3):215–232. 390. Rorstad O. Prognostic indicators for carcinoid
350. Mulligan LM, Marsh DJ, Robinson BG. Genotype- 372. Nuttall KL, Pingree SS. The incidence of elevations neuroendocrine tumors of the gastrointestinal tract.
phenotype correlation in multiple endocrine neopla- in urine 5-hydroxyindoleacetic acid. Ann Clin Lab J Clin Oncol. 2005;89:151–160.
sia type 2: report of the International Consortium. Sci. 1998;28:167–174. 391. Richter G, Stockmann F, Conlon JM, Creutzfeldt
J Intern Med. 1995;238:343–346. 373. Feldman JM, Lee EM. Serotonin content of foods: W. Serotonin release into blood after food and pen-
351. Hofstra RM, Landsvater RM, Ceccherini I. A muta- effect on urinary excretion of 5-hydroxyindoleacetic tagastrin. Studies in healthy subjects and in patients
tion in the RET proto-oncogene associated with acid. Am JClin Nutr. 1985;42:639–643. with metastatic carcinoid tumors. Gastroenterology.
multiple endocrine neoplasia type 2B and sporadic 374. Moertel CG. Karnofsky Memorial Lecture. An 1986;91:612–618.
medullary thyroid carcinoma. Nature. 1994;367: odyssey in the land of small tumors. J Clin Oncol. 392. Adamson AR, Grahame-Smith DG, Peart WS, Starr
375–376. 1987;5:1502–1522. M. Pharmacological blockade of carcinoid flushing
352. Kloos RT, Eng C, Evans DB. Medullary thyroid 375. Tellez MR, Mamikunian G, O’Dorisio TM, Vinik provoked by catecholamines and alcohol. Lancet.
cancer: management guidelines of the American AI, Woltering EA. A single fasting plasma 5-HIAA 1969;2:293–297.
Thyroid Association. Thyroid. 2009;19:565–612. value correlates with 24-hour urinary 5-HIAA values 393. Robertson JI, Peart WS, Andrews TM. The mecha-
353. Wells SA Jr, Dilley WG, Farndon JA, Leight GS, and other biomarkers in midgut neuroendocrine nism of facial flushes in the carcinoid syndrome. Q
Baylin SB. Early diagnosis and treatment of medul- tumors (NETs). Pancreas. 2013;42:405–410. J Med. 1962;31:103–123.
lary thyroid carcinoma. Arch Intern Med. 1985;145: 376. Ardill JE, Armstrong L, Smye M, Doherty R, 394. Moertel CG. Treatment of the carcinoid tumor and
1248–1252. McCance DR, Johnston BT. Neuroendocrine the malignant carcinoid syndrome. J Clin Oncol.
354. Cupisti K, Wolf A, Raffel A, Schott M, Miersch D, tumours of the small bowel: interpretation of raised 1983;1:727–740.
Yang Q, et al. Long-term clinical and biochemical fol- circulating chromogranin A, urinary 5 hydroxy 395. Roberts LJ 2nd, Marney SR Jr, Oates JA. Blockade of
low-up in medullary thyroid carcinoma: a single insti- indole acetic acid and circulating neurokinin A. the flush associated with metastatic gastric carcinoid
tution’s experience over 20 years. Ann Surg. 2007;246: QJM. 2016;109:111–115. by combined histamine H1 and H2 receptor antago-
815–821. 377. Oberg K. Circulating biomarkers in gastroentero- nists. Evidence for an important role of H2 receptors
355. Miccoli P, Minuto MN, Ugolini C, Molinaro E, pancreatic neuroendocrine tumours. Endocr Relat in human vasculature. N Engl J Med. 1979;300:
Basolo F, Berti P, et al. Clinically unpredictable prog- Cancer. 2011;18(suppl 1):S17–S25. 236–238.
nostic factors in the outcome of medullary thyroid 378. Zatelli MC, Torta M, Leon A, Ambrosio MR, Gion 396. Bhattacharyya S, Toumpanakis C, Caplin ME, Davar
cancer. Endocr Relat Cancer. 2007;14:1099–1105. M, Tomassetti P, et al. Chromogranin A as a marker J. Analysis of 150 patients with carcinoid syndrome
356. Baloch ZW, LiVolsi VA. Prognostic factors in of neuroendocrine neoplasia: an Italian Multicenter seen in a single year at one institution in the first
well-differentiated follicular-derived carcinoma and Study. Endocr Relat Cancer. 2007;14:473–482. decade of the twenty-first century. Am J Cardiol.
medullary thyroid carcinoma. Thyroid. 2001;11: 379. Campana D, Nori F, Piscitelli L, Morselli-Labate 2008;101:378–381.
637–645. AM, Pezzilli R, Corinaldesi R, et al. Chromogranin 397. Roberts WC, Sjoerdsma A. The cardiac disease
357. Yip L, Cote GJ, Shapiro SE. Multiple endocrine neo- A: is it a useful marker of neuroendocrine tumors? associated with the carcinoid syndrome (carcinoid
plasia type 2: evaluation of the genotype-phenotype J Clin Oncol. 2007;25:1967–1973. heart disease). Am J Med. 1964;36:5–34.
relationship. Arch Surg. 2003;138:409–416, discus- 380. Reidy-Lagunes DL, Gollub MJ, Saltz LB. Addition 398. Lundin L. Carcinoid heart disease. A cardiologists
sion 416. of octreotide functional imaging to cross-sectional viewpoint. Acta Oncol. 1991;30:499–502.
358. Yip L, Lee JE, Shapiro SE. Surgical management computed tomography or magnetic resonance 399. Bhattacharyya S, Toumpanakis C, Caplin ME, Davar
of hereditary pheochromocytoma. J Am Coll Surg. imaging for the detection of neuroendocrine tumors: J. Usefulness of N-terminal pro-brain natriuretic
2004;198:525–534, discussion 534–535. added value or an anachronism? J Clin Oncol. 2011; peptide as a biomarker of the presence of carcinoid
359. Lee JE, Curley SA, Gagel RF. Cortical-sparing 29:e74–e75. heart disease. Am J Cardiol. 2008;102:938–942.
adrenalectomy for patients with bilateral pheochro- 381. Ganeshan D, Bhosale P, Yang T, Kundra V. Imaging 400. Denney WD, Kemp WE Jr, Anthony LB.
mocytoma. Surgery. 1996;120:1064–1070, discussion features of carcinoid tumors of the gastrointestinal Echocardiographic and biochemical evaluation
1070–1071. tract. AJR Am J Roentgenol. 2013;201:773–786. of the development and progression of carcinoid
360. Obendorfer S. Karzinoide tumoren des dunndarms. 382. Dromain C, de Baere T, Lumbroso J, Caillet H, heart disease. J Am Coll Cardiol. 1998;32:1017–
Frankf Z Pathol. 1907;1:426–430. Laplanche A, Boige V, et al. Detection of liver 1022.
361. Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, metastases from endocrine tumors: a prospective 401. Korse CM, Taal BG, de Groot CA. Chromogranin-A
Mares JE, et al. One hundred years after “carcinoid”: comparison of somatostatin receptor scintigraphy, and N-terminal pro-brain natriuretic peptide: an
epidemiology of and prognostic factors for neuro- computed tomography, and magnetic resonance excellent pair of biomarkers for diagnostics in
endocrine tumors in 35,825 cases in the United imaging. J Clin Oncol. 2005;23:70–78. patients with neuroendocrine tumor. J Clin Oncol.
States. J Clin Oncol. 2008;26:3063–3072. 383. Sippel RS, Chen H. Carcinoid tumors. Surg Oncol 2009;27:4293–4299.
362. Modlin IM, Lye KD, Kidd M. A 5-decade analysis of Clin. 2006;15:463–478. 402. Thompson GB, van Heerden JA Jr, Martin JK.
13,715 carcinoid tumors. Cancer. 2003;97:934–959. 384. Kvols LK, Brown ML, Connor MK. Evaluation of a Carcinoid tumors of the gastrointestinal tract:
363. Alessandro M, Mariani P, Lomanto D. Serum radiolabeled somatostatin analog (I-123 octreotide) presentation, management, and prognosis. Surgery.
neuron-specific enolase in diagnosis and follow-up in the detection and localization of carcinoid 1985;98:1054–1063.
of gastrointestinal neuroendocrine tumors. Tumour and islet cell tumors. Radiology. 1993;187:129– 403. Moertel CG, Dockerty MB, Judd ES. Carcinoid
Biol. 1992;13(5–6):352–357. 133. tumors of the vermiform appendix. Cancer. 1968;21:
364. Schurmann G, Raeth U, Wiedenmann B. Serum 385. Sadowski SM, Neychev V, Millo C, Shih J, 270–278.
chromogranin A in the diagnosis and follow-up of Nilubol N, Herscovitch P, et  al. Prospective 404. Pape UF, Perren A, Niederle B, Gross D, Gress
neuroendocrine tumors of the gastroenteropancreatic study of 68Ga-DOTATATE positron emission T, Costa F, et al. ENETS consensus guidelines for
tract. World J Surg. 1992;16:697–701, discussion tomography/computed tomography for detecting the management of patients with neuroendocrine
701–702. gastro-entero-pancreatic neuroendocrine tumors and neoplasms from the jejuno-ileum and the appendix
365. Rindi G. The E. guidelines: the new TNM clas- unknown primary sites. J Clin Oncol. 2016;34:588– including goblet cell carcinomas. Neuroendocrinology.
sification system. Tumori. 2010;96:806–809. 596. 2012;95:135–156.
1107.e8 Part III: Specific Malignancies

405. Chen T, Yao LQ, Xu MD, Zhang YQ, Chen WF, neuroendocrine midgut tumors: a report from the 439. Gupta S, Johnson MM, Murthy R. Hepatic arterial
Shi Q, et al. Efficacy and safety of endoscopic Group. J Clin Oncol. 2009;27:4656–4663. embolization and chemoembolization for the treat-
submucosal dissection for colorectal carcinoids. 423. Caplin ME, Pavel M, Cwikla JB, Phan AT, Raderer ment of patients with metastatic neuroendocrine
Clin Gastroenterol Hepatol. 2016;14:575–581. M, Sedlackova E, et al. Lanreotide in metastatic tumors: variables affecting response rates and
406. Pinchot SN, Holen K, Sippel RS, Chen H. Carcinoid enteropancreatic neuroendocrine tumors. N Engl survival. Cancer. 2005;104:1590–1602.
tumors. Oncologist. 2008;13:1255–1269. J Med. 2014;371:224–233. 440. Roche A, Girish BV, de Baere T. Prognostic factors
407. Kim MK, Warner RR, Roayaie S, Harpaz N, Ward 424. Rinke A, Wittenberg M, Schade-Brittinger C, for chemoembolization in liver metastasis from
SC, Itzkowitz S, et al. Revised staging classification Aminossadati B, Ronicke E, Gress TM, et al. endocrine tumors. Hepatogastroenterology. 2004;51:
improves outcome prediction for small intestinal Placebo-controlled, double-blind, prospective, 1751–1756.
neuroendocrine tumors. J Clin Oncol. 2013;31: randomized study on the effect of octreotide LAR 441. King J, Glenn D, Quinn R. Radioembolization with
3776–3781. in the control of tumor growth in patients with meta- selective internal radiation microspheres for neuroen-
408. Edge SB, Byrd DR, Compton CC, New S, et al, static neuroendocrine midgut tumors (PROMID): docrine liver metastases. Cancer. 2008;113:921–929.
eds. AJCC (American Joint Committee on Cancer) results of long-term survival. Neuroendocrinology. 442. Henn AR, Levine EA, McNulty W, Zagoria RJ.
Cancer Staging Manual. 7th ed. 2010:181. 2017;104:26–32. Percutaneous radiofrequency ablation of hepatic
409. Strosberg J, Gardner N, Kvols L. Survival and 425. Trendle MC, Moertel CG, Kvols LK. Incidence metastases for symptomatic relief of neuroendo-
prognostic factor analysis of 146 metastatic neuroen- and morbidity of cholelithiasis in patients receiving crine syndromes. AJR Am J Roentgenol. 2003;181:
docrine tumors of the mid-gut. Neuroendocrinology. chronic octreotide for metastatic carcinoid and malig- 1005–1010.
2009;89:471–476. nant islet cell tumors. Cancer. 1997;79:830–834. 443. Gamblin TC, Christians K, Pappas SG. Radio-
410. Jann H, Roll S, Couvelard A, Hentic O, Pavel M, 426. Jayakrishnan TT, Groeschl RT, George B, Thomas frequency ablation of neuroendocrine hepatic
Muller-Nordhorn J, et al. Neuroendocrine tumors of JP, Clark Gamblin T, Turaga KK. Review of the metastasis. Surg Oncol Clin N Am. 2011;20:273–279,
midgut and hindgut origin: tumor-node-metastasis impact of antineoplastic therapies on the risk for vii–viii.
classification determines clinical outcome. Cancer. cholelithiasis and acute cholecystitis. Ann Surg Oncol. 444. Moreno A, Akcakanat A, Munsell MF. Antitumor
2011;117:3332–3341. 2014;21:240–247. activity of rapamycin and octreotide as single agents
411. Landerholm K, Zar N, Andersson RE, Falkmer 427. Attanasio R, Mainolfi A, Grimaldi F, Cozzi R, or in combination in neuroendocrine tumors. Endocr
SE, Jarhult J. Survival and prognostic factors in Montini M, Carzaniga C, et al. Somatostatin Relat Cancer. 2008;15:257–266.
patients with small bowel carcinoid tumour. Br J analogs and gallstones: a retrospective survey on 445. Yao JC, Shah MH, Ito T. Everolimus for advanced
Surg. 2011;98:1617–1624. a large series of acromegalic patients. J Endocrinol pancreatic neuroendocrine tumors. N Engl J Med.
412. Chen H, Hardacre JM, Uzar A. Isolated liver Invest. 2008;31:704–710. 2011;364:514–523.
metastases from neuroendocrine tumors: does resec- 428. Sundstrom S, Bremnes RM, Kaasa S. Cisplatin and 446. Yao JC, Fazio N, Singh S, Buzzoni R, Carnaghi
tion prolong survival? Coll Surg. 1998;187:88–92, etoposide regimen is superior to cyclophosphamide, C, Wolin E, et al. Everolimus for the treatment of
discussion 92–93. epirubicin, and vincristine regimen in small-cell lung advanced, non-functional neuroendocrine tumours
413. Mayo SC, de Jong MC, Pulitano C, Clary BM, cancer: results from a randomized phase III trial with of the lung or gastrointestinal tract (RADIANT-4):
Reddy SK, Gamblin TC, et al. Surgical management 5 years’ follow-up. J Clin Oncol. 2002;20:4665–4672. a randomised, placebo-controlled, phase 3 study.
of hepatic neuroendocrine tumor metastasis: results 429. Iwasa S, Morizane C, Okusaka T. Cisplatin and Lancet. 2016;387:968–977.
from an international multi-institutional analysis. etoposide as first-line chemotherapy for poorly 447. Pavel ME, Hainsworth JD, Baudin E. Everolimus
Ann Surg Oncol. 2010;17:3129–3136. differentiated neuroendocrine carcinoma of the plus octreotide long-acting repeatable for the
414. Que FG, Sarmiento JM, Nagorney DM. Hepatic hepatobiliary tract and pancreas. Jpn J Clin Oncol. treatment of advanced neuroendocrine tumours
surgery for metastatic gastrointestinal neuroendocrine 2010;40:313–318. associated with carcinoid syndrome (RADIANT-2):
tumors. Cancer Control. 2002;9:67–79. 430. Moertel CG, Hanley JA. Combination chemo- a randomised, placebo-controlled, phase 3 study.
414a. Mazzaferro V, Sposito C, Coppa J, et al. The therapy trials in metastatic carcinoid tumor and Lancet. 2011;378:2005–2012.
long-term benefit of liver transplantation for the malignant carcinoid syndrome. Cancer Clin 448. Yao JC, Phan A, Hoff PM. Targeting vascular
hepatic metastases from neuroendocrine tumors. Trials. 1979;2:327–334. endothelial growth factor in advanced carcinoid
Am J Transplant. 2016;16(10):2892–2902. 431. Sun W, Lipsitz S, Catalano P. Phase II/III study tumor: a random assignment phase II study of depot
414b. Rossi RE, Burroughs AK, Caplin ME. Liver of doxorubicin with fluorouracil compared with octreotide with bevacizumab and pegylated interferon
transplantation for unresectable neuroendocrine streptozocin with fluorouracil or dacarbazine in the alpha-2b. J Clin Oncol. 2008;26:1316–1323.
tumor liver metastases. Ann Surg Oncol. 2014;21(7): treatment of advanced carcinoid tumors: Eastern 449. Raymond E, Dahan L, Raoul JL. Sunitinib malate for
2398–2405. Cooperative Oncology Group Study E1281. J Clin the treatment of pancreatic neuroendocrine tumors.
415. Rusthoven KE, Kavanagh BD, Cardenes H. Oncol. 2005;23:4897–4904. N Engl J Med. 2011;364:501–513.
Multi-institutional phase I/II trial of stereotactic 432. Oberg K, Eriksson B. Medical treatment of neuro- 450. Kulke MH, Lenz HJ, Meropol NJ. Activity of
body radiation therapy for liver metastases. J Clin endocrine gut and pancreatic tumors. Acta Oncol. sunitinib in patients with advanced neuroendocrine
Oncol. 2009;27:1572–1578. 1989;28:425–431. tumors. J Clin Oncol. 2008;26:3403–3410.
416. Kvols LK, Moertel CG, O’Connell MJ, Schutt AJ, 433. Moertel CG, Rubin J, Kvols LK. Therapy of meta- 451. Cwikla JB, Sankowski A, Seklecka N. Efficacy of
Rubin J, Hahn RG. Treatment of the malignant static carcinoid tumor and the malignant carcinoid radionuclide treatment DOTATATE Y-90 in patients
carcinoid syndrome. Evaluation of a long-acting syndrome with recombinant leukocyte A interferon. with progressive metastatic gastroenteropancreatic
somatostatin analogue. N Engl J Med. 1986;315: J Clin Oncol. 1989;7:865–868. neuroendocrine carcinomas (GEP-NETs): a phase
663–666. 434. Tiensuu Janson EM, Ahlstrom H, Andersson T, II study. Ann Oncol. 2010;21:787–794.
417. Frolich JC, Bloomgarden ZT, Oates JA. The carcinoid Oberg KE. Octreotide and interferon alfa: a new 452. Kaltsas GA, Papadogias D, Makras P, Grossman
flush. Provocation by pentagastrin and inhibition by combination for the treatment of malignant carcinoid AB. Treatment of advanced neuroendocrine tumours
somatostatin. N Engl J Med. 1978;299:1055–1057. tumours. Eur J Cancer. 1992;28a:1647–1650. with radiolabelled somatostatin analogues. Endocr
418. Rubin J, Ajani J, Schirmer W. Octreotide acetate 435. Oberg K, Eriksson B, Janson ET. Interferons alone Relat Cancer. 2005;12:683–699.
long-acting formulation versus open-label subcu- or in combination with chemotherapy or other 453. Strosberg JR, Wolin EM, Chasen B, Lu D.
taneous octreotide acetate in malignant carcinoid biologicals in the treatment of neuroendocrine gut NETTER-1 phase III: efficacy and safety results
syndrome. J Clin Oncol. 1999;17:600–606. and pancreatic tumors. Digestion. 1994;55(suppl 3): in patients with midgut neuroendocrine tumors
419. Grosman I, Simon D. Potential gastrointestinal uses 64–69. treated with 177(abstract). J Clin Oncol. 2016;34.
of somatostatin and its synthetic analogue octreotide. 436. Pavel M, Baudin E, Couvelard A, Krenning E, 454. Villard L, Romer A, Marincek N. Cohort study
Am J Gastroenterol. 1990;85:1061–1072. Oberg K, Steinmuller T, et al. ENETS Consensus of somatostatin-BASED radiopeptide therapy with
420. Kvols LK, Moertel CG, Connell MJ. Treatment of Guidelines for the management of patients with liver [90Y-DOTA]-[90Y-DOTA]-[177Lu-DOTA]-TOC
the malignant carcinoid syndrome. Evaluation of a and other distant metastases from neuroendocrine in neuroendocrine cancers. J Clin Oncol. 2012;30:
longacting somatostatin analogue. N Engl J Med. neoplasms of foregut, midgut, hindgut, and unknown 1100–1106.
1986;315:663–666. primary. Neuroendocrinology. 2012;95:157–176. 455. Hallet J, Law CH, Cukier M, Saskin R, Liu N, Singh
421. Wynick D, Anderson JV, Williams SJ, Bloom 437. Kunz PL, Reidy-Lagunes D, Anthony LB, Bertino S. Exploring the rising incidence of neuroendocrine
SR. Resistance of metastatic pancreatic endocrine EM, Brendtro K, Chan JA, et al. Consensus tumors: a population-based analysis of epidemiology,
tumours after long-term treatment with the soma- guidelines for the management and treatment of metastatic presentation, and outcomes. Cancer.
tostatin analogue octreotide (SMS). Clin Endocrinol neuroendocrine tumors. Pancreas. 2013;42:557–577. 2015;121:589–597.
(Oxf ). 1989;30:201–995, 385–388. 438. Vogl TJ, Naguib NNN, Zangos S. Liver metastases 456. Harris AG, Dorisio TM, Woltering EA. Consensus
422. Rinke A, Muller HH, Schade-Brittinger C. Placebo- of neuroendocrine carcinomas: interventional statement: octreotide dose titration in secretory diar-
controlled, double-blind, prospective, randomized treatment via transarterial embolization, chemoem- rhea. Diarrhea Management Consensus Development
study on the effect of octreotide LAR in the bolization and thermal ablation. Eur J Endocrinol Panel Digestive diseases and sciences. Dig Dis Sci.
control of tumor growth in patients with metastatic 2009;72:517–528. 1995;40:1464–1473.
Cancer of the Endocrine System  •  CHAPTER 68 1107.e9
1107.e9

457. Leoncini E, Carioli G, La Vecchia C, Boccia S, 478. Bone HG 3rd. Diagnosis of the multiglandular 500. Bloom SR, Polak JM. Glucagonoma syndrome. Am
Rindi G. Risk factors for neuroendocrine neoplasms: endocrine neoplasias. Clin Chem. 1990;36:711–718. J Surg. 1987;82:25–36.
a systematic review and meta-analysis. Ann Oncol. 479. Oberg K, Eriksson B. Endocrine tumours of 501. Chu QD, Al-kasspooles MF, Smith JL, Nava HR,
2016;27:68–81. the pancreas. Best Pract Res Clin Gastroenterol. Douglass HO Jr, Driscoll D, et al. Is glucagonoma
458. Haugvik SP, Hedenstrom P, Korsaeth E, Valente R, 2005;19:753–781. of the pancreas a curable disease? Int J Pancreatol.
Hayes A, Siuka D, et al. Diabetes, smoking, alcohol 480. Ayav A, Bresler L, Brunaud L, Boissel P. Laparoscopic 2001;29:155–162.
use, and family history of cancer as risk factors for approach for solitary insulinoma: a multicentre study. 502. Krejs GJ, Orci L, Conlon JM. Somatostatinoma
pancreatic neuroendocrine tumors: a systematic review Langenbecks Arch Surg. 2005;390:134–140. syndrome. Biochemical morphologic and clinical
and meta-analysis. Neuroendocrinology. 2015;101: 481. Finlayson E, Clark OH. Surgical treatment of features. N Engl J Med. 1979;301:285–292.
133–142. insulinomas. Surg Clin North Am. 2004;84:775–785. 503. Harris GJ, Tio F, Cruz AB Jr. Somatostatinoma:
459. Legmann P, Vignaux O, Dousset B, Baraza AJ, Palazzo 482. Power L. A glucose-responsive insulinoma. JAMA. a case report and review of the literature. J Surg
L, Dumontier I, et al. Pancreatic tumors: comparison 1969;207:893–896. Oncol. 1987;36:8–16.
of dual-phase helical CT and endoscopic sonography. 483. Maton PN. The use of the long-acting somatostatin 504. Doherty GM. Rare endocrine tumours of the GI
AJR Am J Roentgenol. 1998;170:1315–1322. analogue, octreotide acetate, in patients with islet tract. Best Pract Res Clin Gastroenterol. 2005;19:807–
460. Khashab MA, Yong E, Lennon AM, Shin EJ, cell tumors. Gastroenterol Clin North Am. 1989;18: 817.
Amateau S, Hruban RH, et al. EUS is still superior 897–922. 505. Kulke MH, Shah MH, Benson AB 3rd, Bergsland
to multidetector computerized tomography for 484. Holland J, Bast R, Morton D, eds. Cancer Medicine. E, Berlin JD, Blaszkowsky LS, et al. Neuroendocrine
detection of pancreatic neuroendocrine tumors. 1997. tumors, version 1.2015. J Natl Compr Canc Netw.
Gastrointest Endosc. 2011;73:691–696. 485. Roth H, Thier S, Segal S. Zinc glucagon in the 2015;13:78–108.
461. Gouya H, Vignaux O, Augui J. CT, endoscopic management of refractory hypoglycemia due to 506. Soga J, Yakuwa Y. Somatostatinoma/inhibitory
sonography, and a combined protocol for preopera- insulin-producing tumors. N Engl J Med. 1966;274: syndrome: a statistical evaluation of 173 reported
tive evaluation of pancreatic insulinomas. AJR Am 493–497. cases as compared to other pancreatic endocrinomas.
J Roentgenol. 2003;181:987–992. 486. Landau BR, Levine HJ, Hertz R. Prolonged glucagon J Exp Clin Cancer Res. 1999;18:13–22.
462. Ichikawa T, Peterson MS, Federle MP. Islet cell tumor administration in a case of hyperinsulinism due to 507. Norton JA. Neuroendocrine tumors of the pancreas
of the pancreas: biphasic CT versus MR imaging in disseminated islet cell carcinoma. N Engl J Med. and duodenum. Curr Probl Surg. 1994;31:77–156.
tumor detection. Radiology. 2000;216:163–171. 1958;259:286–288. 508. Jensen RT, Gardner JD, Raufman JP. Zollinger-
463. Thoeni RF, Mueller-Lisse UG, Chan R. Detection 487. Graber AL Jr, Williams D, Porte RH. Clinical use Ellison syndrome: current concepts and manage-
of small, functional islet cell tumors in the pancreas: of diazoxide and mechanism for its hyperglycemic ment. Ann Intern Med. 1983;98:59–75.
selection of MR imaging sequences for optimal effects. Diabetes. 1966;15:143–148. 509. Levy AD, Sobin LH. From the archives of the
sensitivity. Radiology. 2000;214:483–490. 488. Hirshberg B, Cochran C, Skarulis MC, Libutti AFIP: gastrointestinal carcinoids: imaging features
464. Owen NJ, Sohaib SA, Peppercorn PD. MRI of SK, Alexander HR, Wood BJ, et al. Malignant with clinicopathologic comparison. Radiographics.
pancreatic neuroendocrine tumours. Br J Radiol. insulinoma: spectrum of unusual clinical features. 2007;27:237–257.
2001;74:968–973. Cancer. 2005;104:264–272. 510. Norton JA, Alexander HR, Fraker DL, Venzon
465. Semelka RC, Custodio CM, Cem Balci N, Woosley 489. Jensen RT, Cadiot G, Brandi ML, de Herder WW, DJ, Gibril F, Jensen RT. Possible primary lymph
JT. Neuroendocrine tumors of the pancreas: spec- Kaltsas G, Komminoth P, et al. ENETS Consensus node gastrinoma: occurrence, natural history, and
trum of appearances on MRI. J Magn Reson Imaging. Guidelines for the management of patients with predictive factors: a prospective study. Ann Surg.
2000;11:141–148. digestive neuroendocrine neoplasms: functional 2003;237:650–657, discussion 7–9.
466. Chang F, Chandra A, Culora G. Cytologic diagnosis pancreatic endocrine tumor syndromes. Neuroen- 511. Ellison EC, Carey LC, Sparks J. Early surgical
of pancreatic endocrine tumors by endoscopic docrinology. 2012;95:98–119. treatment of gastrinoma. Am J Surg. 1987;82:17–24.
ultrasound-guided fine-needle aspiration: a review. 490. Wermers RA, Fatourechi V, Wynne AG, Kvols LK, 512. McGuigan JE, Wolfe MM. Secretin injection test
Diagn Cytopathol. 2006;34:649–658. Lloyd RV. The glucagonoma syndrome. Clinical in the diagnosis of gastrinoma. Gastroenterology.
467. Rosch T, Lightdale CJ, Botet JF. Localization of and pathologic features in 21 patients. Medicine 1980;79:1324–1331.
pancreatic endocrine tumors by endoscopic ultra- (Baltimore). 1996;75:53–63. 513. Berna MJ, Hoffmann KM, Long SH, Serrano J,
sonography. N Engl J Med. 1992;326:1721-1726. 491. Friesen SR. Tumors of the endocrine pancreas. N Gibril F, Jensen RT. Serum gastrin in Zollinger-
468. Okabayashi T, Shima Y, Sumiyoshi T, Kozuki A, Ito Engl J Med. 1982;306:580–590. Ellison syndrome: II. Prospective study of gastrin
S, Ogawa Y, et al. Diagnosis and management of 492. Stacpoole PW. The glucagonoma syndrome: clini- provocative testing in 293 patients from the National
insulinoma. World J Gastroenterol. 2013;19:829–837. cal features, diagnosis, and treatment. Endocr Rev. Institutes of Health and comparison with 537 cases
469. Placzkowski KA, Vella A, Thompson GB, Grant CS, 1981;2:347–361. from the literature. evaluation of diagnostic criteria,
Reading CC, Charboneau JW, et al. Secular trends 493. Levy-Bohbot N, Merle C, Goudet P, Delemer B, proposal of new criteria, and correlations with
in the presentation and management of functioning Calender A, Jolly D, et al. Prevalence, characteristics clinical and tumoral features. Medicine (Baltimore).
insulinoma at the Mayo Clinic, 1987-2007. J Clin and prognosis of MEN 1-associated glucagonomas, 2006;85:331–364.
Endocrinol Metab. 2009;94:1069–1073. VIPomas, and somatostatinomas: study from 514. Meko JB, Norton JA. Management of patients
470. Service FJ, McMahon MM, O’Brien PC, Ballard the GTE (Groupe des Tumeurs Endocrines) with Zollinger-Ellison syndrome. Annu Rev Med.
DJ. Functioning insulinoma–incidence, recurrence, registry. Gastroenterol Clin Biol. 2004;28:1075– 1995;46:395–411.
and long-term survival of patients: a 60-year study. 1081. 515. O’Toole D, Grossman A, Gross D. ENETS Guide-
Mayo Clin Proc. 1991;66:711–719. 494. Castro PG, de Leon AM, Trancon JG, Martinez PA, lines for the Standards of Care in Neuroendocrine
471. Fajans SS, Vinik AI. Insulin-producing islet cell Alvarez Perez JA, Fernandez Fernandez JC, et al. Tumors: biochemical markers. Neuroendocrinology.
tumors. Endocrinol Metab Clin North Am. 1989;18: Glucagonoma syndrome: a case report. J Med Case 2009;90:194–202.
45–74. Rep. 2011;5:402. 516. Gibril F, Reynolds JC, Doppman JL. Somatostatin
472. Boden G. Insulinoma and glucagonoma. Semin 495. Higgins GA, Recant L, Fischman AB. The gluca- receptor scintigraphy: its sensitivity compared with
Oncol. 1987;14:253–262. gonoma syndrome: surgically curable diabetes. Am that of other imaging methods in detecting primary
473. Grunberger G, Weiner JL, Silverman R. Factitious J Surg. 1979;137:142–148. and metastatic gastrinomas. A prospective study.
hypoglycemia due to surreptitious administration of 496. Weinel RJ, Neuhaus C, Stapp J. Preoperative Ann Intern Med. 1996;125:26–34.
insulin. Diagnosis treatment and longterm followup. localization of gastrointestinal endocrine tumors 517. Zollinger RM, Ellison EC, Fabri PJ. Primary peptic
Ann Intern Med. 1988;108:252–257. using somatostatin-receptor scintigraphy. Ann Surg. ulcerations of the jejunum associated with islet
474. Ferrer-Garcia JC, Iranzo Gonzalez-Cruz V, Navas- 1993;218:640–645. cell tumors. Twenty-five year appraisal. Ann Surg.
DeSolis S, Civera-Andres M, Morillas-Arino C, 497. Kirkwood KS, Debas HT. Neuroendocrine tumors: 1980;192:422–430.
Merchante-Alfaro A, et al. Management of malignant common presentations of uncommon diseases. 518. Bonfils S, Landor JH, Mignon M, Hervoir P.
insulinoma. Clin Transl Oncol. 2013;15:725–731. Compr Ther. 1995;21:719–725. Results of surgical management in 92 consecutive
475. Tucker ON, Crotty PL, Conlon KC. The manage- 498. Poultsides GA, Huang LC, Chen Y, Visser BC, patients with Zollinger-Ellison syndrome. Ann Surg.
ment of insulinoma. Br J Surg. 2006;93:264–275. Pai RK, Jeffrey RB, et al. Pancreatic neuroen- 1981;194:692–697.
476. Kahn CR, Rosen SW, Weintraub BD. Ectopic docrine tumors: radiographic calcifications cor- 519. Metz DC, Comer GM, Soffer E. Three-year
production of chorionic gonadotropin and its relate with grade and metastasis. Ann Surg Oncol. oral pantoprazole administration is effective for
subunits by islet-cell tumors. A specific marker for 2012;19:2295–2303. patients with Zollinger-Ellison syndrome and other
malignancy. N Engl J Med. 1977;297:565–569. 499. Frilling A, Li J, Malamutmann E, Schmid KW, hypersecretory conditions. Aliment Pharmacol Ther.
477. Alsever RN, Roberts JP, Gerber JG. Insulinoma Bockisch A, Broelsch CE. Treatment of liver 2006;23:437–444.
with low circulating insulin levels: the diagnostic metastases from neuroendocrine tumours in rela- 520. Hirschowitz BI, Simmons J, Mohnen J. Clinical
value of proinsulin measurements. Ann Intern Med. tion to the extent of hepatic disease. Br J Surg. outcome using lansoprazole in acid hypersecretors
1975;82:347–350. 2009;96:175–184. with and without Zollinger-Ellison syndrome: a
1107.e10 Part III: Specific Malignancies

13-year prospective study. Clin Gastroenterol Hepatol. 539. Kulke MH, Niedzwiecki D, Foster NR. Randomized 559. Messerer CL, Bugis SP, Baliski C, Wiseman SM.
2005;3:39–48. phase II study of everolimus versus everolimus plus Normocalcemic parathyroid carcinoma: an unusual
521. Norton JA, Fraker DL, Alexander HR, Venzon bevacizumab in patients with locally advanced or clinical presentation. World J Surg Oncol. 2006;4:10.
DJ, Doppman JL, Serrano J, et al. Surgery to cure metastatic pancreatic neuroendocrine tumors 560. Sahasranam P, Tran MT, Mohamed H, Friedman
the Zollinger-Ellison syndrome. N Engl J Med. (pNET),CALGB 80701 (Alliance) (abstr). Oncol TC. Multiglandular parathyroid carcinoma: a case
1999;341:635–644. (suppl; abstr 4005). Abstract available online at report and brief review. South Med J. 2007;100:841–
522. Norton JA, Fraker DL, Alexander HR, Gibril F, http://meetinglibrary.asco.org/content/. Accessed 844.
Liewehr DJ, Venzon DJ, et al. Surgery increases sur- on September 4, 2015. 561. Busaidy NL, Jimenez C, Habra MA. Parathyroid car-
vival in patients with gastrinoma. Ann Surg. 2006;244: 540. Dudney WC, Bodenner D, Stack BC Jr. Parathyroid cinoma: a 22-year experience. Head Neck. 2004;26:
410–419. carcinoma. Otolaryngol Clin North Am. 2010;43: 716–726.
522a. Yu F, Venzon DJ, Serrano J, et al. Prospective 441–453, xi. 562. Kebebew E. Parathyroid carcinoma. Curr Treat
study of the clinical course, prognostic factors, 541. Sandelin K, Auer G, Bondeson L, Grimelius L, Options Oncol. 2001;2:347–354.
causes of death, and survival in patients with long- Farnebo LO. Prognostic factors in parathyroid cancer: 563. Shane E. Clinical review 122: parathyroid carcinoma.
standing Zollinger-Ellison syndrome. J Clin Oncol. a review of 95 cases. World J Surg. 1992;16:724–731. J Clin Endocrinol Metab. 2001;86:485–493.
1999;17(2):615–630. 542. Kebebew E, Arici C, Duh QY, Clark OH. 564. Hara H, Igarashi A, Yano Y, Yashiro T, Ueno
523. Verner JV, Morrison AB. Islet cell tumor and a Localization and reoperation results for persistent E, Aiyoshi Y, et al. Ultrasonographic features of
syndrome of refractory watery diarrhea and hypo- and recurrent parathyroid carcinoma. Arch Surg. parathyroid carcinoma. Endocr J. 2001;48:213–217.
kalemia. Am J Surg. 1958;25:374–380. 2001;136:878–885. 565. Sidhu PS, Talat N, Patel P, Mulholland NJ, Schulte
524. Kane MG, Dorisio TM, Krejs GJ. Production of 543. Lee PK, Jarosek SL, Virnig BA. Trends in the KM. Ultrasound features of malignancy in the
secretory diarrhea by intravenous infusion of vasoac- incidence and treatment of parathyroid cancer preoperative diagnosis of parathyroid cancer: a
tive intestinal polypeptide. N Engl J Med. 1983;309: in the United States. Cancer. 2007;109:1736– retrospective analysis of parathyroid tumours larger
1482–1485. 1741. than 15 mm. Eur Radiol. 2011;21:1865–1873.
525. Soga J, Yakuwa Y. Vipoma/diarrheogenic syndrome: 544. James BC, Aschebrook-Kilfoy B, Cipriani N, Kaplan 566. Gao WC, Ruan CP, Zhang JC, Liu HM, Xu XY,
a statistical evaluation of 241 reported cases. J Exp EL, Angelos P, Grogan RH. The incidence and Sun YP, et al. Nonfunctional parathyroid carcinoma.
Clin Cancer Res. 1998;17:389–400. survival of rare cancers of the thyroid, parathyroid, J Cancer Res Clin Oncol. 2010;136:969–974.
526. Contessa JN, Griffith KA, Wolff E, Ensminger W, adrenal, and pancreas. Ann Surg Oncol. 2016;23: 567. Kassahun WT, Jonas S. Focus on parathyroid
Zalupski M, Lawrence TS, et al. Radiotherapy for 424–433. carcinoma. Int J Surg. 2011;9:13–19.
pancreatic neuroendocrine tumors. Int J Radiat Oncol 545. Arnold A, Shattuck TM, Mallya SM. Molecular 568. Schantz A, Castleman B. Parathyroid carcinoma. A
Biol Phys. 2009;75:1196–1200. pathogenesis of primary hyperparathyroidism. J Bone study of 70 cases. Cancer. 1973;31:600–605.
527. Strosberg J, Hoffe S, Gardner N, Choi J, Kvols L. Miner Res. 2002;17(suppl 2):N30–N36. 569. Bondeson L, Sandelin K, Grimelius L. Histopatho-
Effective treatment of locally advanced endocrine 546. Carpten JD, Robbins CM, Villablanca A. logical variables and DNA cytometry in parathyroid
tumors of the pancreas with chemoradiotherapy. HRPT2, encoding parafibromin, is mutated in carcinoma. Am J Clin Pathol. 1993;17:820–829.
Neuroendocrinology. 2007;85:216–220. hyperparathyroidism-jaw tumor syndrome. Nat 570. Truran PP, Johnson SJ, Bliss RD, Lennard TW,
528. Kvols LK, Buck M, Moertel CG. Treatment of Genet. 2002;32:676–680. Aspinall SR. Parafibromin, galectin-3, PGP9.5, Ki67,
metastatic islet cell carcinoma with a somatostatin 547. Shattuck TM, Valimaki S, Obara T. Somatic and cyclin D1: using an immunohistochemical panel
analogue (SMS). Ann Intern Med. 1987;107:201– and germ-line mutations of the HRPT2 gene in to aid in the diagnosis of parathyroid cancer. World
995, 162–168. sporadic parathyroid carcinoma. N Engl J Med. J Surg. 2014;38:2845–2854.
529. Pavel ME, Baum U, Hahn EG. Efficacy and 2003;349:1722–1729. 571. Wei CH, Harari A. Parathyroid carcinoma: update
tolerability of pegylated IFN-alpha in patients with 548. Howell VM, Haven CJ, Kahnoski K. HRPT2 and guidelines for management. Curr Treat Options
neuroendocrine gastroenteropancreatic carcinomas. mutations are associated with malignancy in spo- Oncol. 2012;13:11–23.
J Interferon Cytokine Res. 2006;26:8–13. radic parathyroid tumours. J Med Genet. 2003;40: 572. Betea D, Potorac I, Beckers A. Parathyroid carcinoma:
530. Schein P, Kahn R, Gorden P. Streptozotocin for 657–663. challenges in diagnosis and treatment. Ann Endocrinol
malignant insulinomas and carcinoid tumor. Report 549. Krebs LJ, Shattuck TM, Arnold A. HRPT2 muta- (Paris). 2015;76:169–177.
of eight cases and review of the literature. Arch Intern tional analysis of typical sporadic parathyroid adeno- 573. Rubin MR, Silverberg SJ, D’Amour P, Brossard JH,
Med. 1973;132:555–561. mas. J Clin Endocrinol Metab. 2005;90:5015–5017. Rousseau L, Sliney J Jr, et al. An N-terminal molecu-
531. Moertel CG, Hanley JA, Johnson LA. Streptozocin 550. Cetani F, Pardi E, Borsari S, Viacava P, Dipollina lar form of parathyroid hormone (PTH) distinct
alone compared with streptozocin plus fluorouracil G, Cianferotti L, et al. Genetic analyses of the from hPTH(1 84) is overproduced in parathyroid
in the treatment of advanced islet-cell carcinoma. HRPT2 gene in primary hyperparathyroidism: carcinoma. Clin Chem. 2007;53:1470–1476.
N Engl J Med. 1980;303:1189–1194. germline and somatic mutations in familial and 574. Caron P, Maiza JC, Renaud C, Cormier C, Barres
532. Cheng PN, Saltz LB. Failure to confirm major objec- sporadic parathyroid tumors. J Clin Endocrinol BH, Souberbielle JC. High third generation/second
tive antitumor activity for streptozocin and doxo- Metab. 2004;89:5583–5591. generation PTH ratio in a patient with parathyroid
rubicin in the treatment of patients with advanced 551. Cryns VL, Thor A, Xu HJ. Loss of the retinoblastoma carcinoma: clinical utility of third generation/second
islet cell carcinoma. Cancer. 1999;86:944–948. tumor-suppressor gene in parathyroid carcinoma. generation PTH ratio in patients with primary hyper-
533. Gonzalez MA, Biswas S, Clifton L, Corrie PG. Treat- N Engl J Med. 1994;330:757–761. parathyroidism. Clin Endocrinol (Oxf ). 2009;70:
ment of neuroendocrine tumours with infusional 552. Cryns VL, Rubio MP, Thor AD. p53 abnormalities 533–538.
5-fluorouracil, folinic acid and streptozocin. Br J in human parathyroid carcinoma. J Clin Endocrinol 575. Cavalier E, Betea D, Schleck ML, Gadisseur R,
Cancer. 2003;89:455–456. Metab. 1994;78:1320–1324. Vroonen L, Delanaye P, et al. The third/second gen-
534. Moertel CG, Lefkopoulo M, Lipsitz S. Streptozocin- 553. Haven CJ, van Puijenbroek M, Tan MH. Identifica- eration PTH assay ratio as a marker for parathyroid
doxorubicin, streptozocin-fluorouracil or chlorozoto- tion of MEN1 and HRPT2 somatic mutations in carcinoma: evaluation using an automated platform.
cin in the treatment of advanced islet-cell carcinoma. paraffin-embedded (sporadic) parathyroid carcino- J Clin Endocrinol Metab. 2014;99:E453–E457.
N Engl J Med. 1992;326:519–523. mas. Clin Endocrinol (Oxf ). 2007;67:370–376. 576. Owen RP, Silver CE, Pellitteri PK. Parathyroid
535. Kouvaraki MA, Ajani JA, Hoff P. Fluorouracil, 554. Singh Ospina N, Sebo TJ, Thompson GB, Clarke carcinoma: a review. Head Neck. 2011;33:429–436.
doxorubicin, and streptozocin in the treatment BL, Young WF Jr. Prevalence of parathyroid car- 577. Shane E, Bilezikian JP. Parathyroid carcinoma: a
of patients with locally advanced and metastatic cinoma in 348 patients with multiple endocrine review of 62 patients. Endocr Rev. 1982;3:218–226.
pancreatic endocrine carcinomas. J Clin Oncol. neoplasia type 1—case report and review of the 578. Sandelin K, Thompson NW, Bondeson L.
2004;22:4762–4771. literature. Clin Endocrinol (Oxf ). 2014. Metastatic parathyroid carcinoma: dilemmas in
536. Kunz PL, Balise RR, Fehrenbacher L, Pan M, Venook 555. Obara T, Fujimoto Y. Diagnosis and treatment of management. Surgery. 1991;110:978–986, discussion
AP, Fisher GA, et al. Oxaliplatin-fluoropyrimidine patients with parathyroid carcinoma: an update and 986–988.
chemotherapy plus bevacizumab in advanced review. World J Surg. 1991;15:738–744. 579. Hoelting T, Weber T, Werner J, Herfarth C. Surgical
neuroendocrine tumors: an analysis of 2 phase II 556. Wynne AG, van Heerden J, Carney JA, Fitzpatrick treatment of parathyroid carcinoma (review). Oncol
trials. Pancreas. 2016;45:1394–1400. LA. Parathyroid carcinoma: clinical and pathologic Rep. 2001;8:931–934.
537. Kulke MH, Stuart K, Enzinger PC. Phase II, study features in 43 patients. Medicine (Baltimore). 580. Harari A, Waring A, Fernandez-Ranvier G, Hwang
of temozolomide and thalidomide in patients with 1992;71:197–205. J, Suh I, Mitmaker E, et al. Parathyroid carcinoma:
metastatic neuroendocrine tumors. J Clin Oncol. 557. Givi B, Shah JP. Parathyroid carcinoma. Clin Oncol a 43-year outcome and survival analysis. J Clin
2006;24:401–406. (R Coll Radiol). 2010;22:498–507. Endocrinol Metab. 2011;96:3679–3686.
538. Strosberg JR, Fine RL, Choi J. First-line che- 558. Wilkins BJ Jr, Lewis JS. Non-functional parathyroid 581. Silverberg SJ, Rubin MR, Faiman C. Cinacalcet
motherapy with capecitabine and temozolomide carcinoma: a review of the literature and report of a hydrochloride reduces the serum calcium concentra-
in patients with metastatic pancreatic endocrine case requiring extensive surgery. Head Neck Pathol. tion in inoperable parathyroid carcinoma. J Clin
carcinomas. Cancer. 2011;117:268–275. 2009;3:140–149. Endocrinol Metab. 2007;92:3803–3808.
Cancer of the Endocrine System  •  CHAPTER 68 1107.e11
1107.e11

582. de Papp AE, Kinder B, LiVolsi V. Parathyroid 587. Karuppiah D, Thanabalasingham G, Shine B, Wang and biochemical normalization and tumor
carcinoma arising from parathyroid hyperplasia: LM, Sadler GP, Karavitaki N, et al. Refractory shrinkage induced by anti-parathyroid hormone
autoinfarction following intravenous treatment hypercalcaemia secondary to parathyroid carcinoma: immunotherapy in a patient with metastatic
with pamidronate. Am J Surg. 1994;97:399–400. response to high-dose denosumab. Eur J Endocrinol. parathyroid carcinoma. J Clin Endocrinol Metab.
583. Szmuilowicz ED, Utiger RD. A case of parathyroid 2014;171:K1–K5. 2004;89:3413–3420.
carcinoma with hypercalcemia responsive to cina- 588. Tochio M, Takaki H, Yamakado K, Uraki J, 592. Simonds WF, Robbins CM, Agarwal SK, Hendy
calcet therapy. Nat Clin Pract Endocrinol Metab. Kashima M, Nakatsuka A, et al. A case report of GN, Carpten JD, Marx SJ. Familial isolated hyper-
2006;2(5):291–296. 20 lung radiofrequency ablation sessions for 50 parathyroidism is rarely caused by germline mutation
584. Vellanki P, Lange K, Elaraj D, Kopp PA, El Muayed lung metastases from parathyroid carcinoma causing in HRPT2, the gene for the hyperparathyroidism-
M. Denosumab for management of parathyroid hyperparathyroidism. Cardiovasc Intervent Radiol. jaw tumor syndrome. J Clin Endocrinol Metab.
carcinoma-mediated hypercalcemia. J Clin Endocrinol 2010;33:657–659. 2004;89:96–102.
Metab. 2014;99:387–390. 589. Artinyan A, Guzman E, Maghami E, Al-Sayed 593. Hundahl SA, Fleming ID, Fremgen AM, Menck
585. Tong CV, Hussein Z, Noor NM, Mohamad M, D’Apuzzo M, Wagman L, et al. Metastatic HR. Two hundred eighty-six cases of parathyroid
M, Ng WF. Use of denosumab in parathyroid parathyroid carcinoma to the liver treated with carcinoma treated in the US between a National
carcinoma with refractory hypercalcemia. QJM. radiofrequency ablation and transcatheter arterial Cancer Data Base Report The American College of
2015;108:49–50. embolization. J Clin Oncol. 2008;26:4039–4041. Surgeons Commission on Cancer and the American
586. Nadarasa K, Theodoraki A, Kurzawinski TR, 590. Bradwell AR, Harvey TC. Control of hypercalcaemia Cancer Society. Cancer. 1999;86:1985–1995,
Carpenter R, Bull J, Chung TT, et al. Denosumab of parathyroid carcinoma by immunisation. Lancet. 538–544.
for management of refractory hypercalcaemia in 1999;353:370–373.
recurrent parathyroid carcinoma. Eur J Endocrinol. 591. Betea D, Bradwell AR, Harvey TC, Mead GP,
2014;171:L7–L8. Schmidt-Gayk H, Ghaye B, et al. Hormonal

You might also like