15
368 Review Article Sandi-Jo Galati, MD*; Meena Said, MD*; Rebekah Gospin, MD; Nikolina Babic, PhD; Karen Brown, MS; Eliza B. Geer, MD; Lale Kostakoglu, MD, MPH; Lawrence R. Krakoff, MD; Andrew B. Leibowitz, MD; Lakshmi Mehta, MD, FACMG; Simone Muller, BS, PharmD, BCPS; Randall P. Owen, MD, MS, FACS; David S. Pertsemlidis, MD, FACS; Eric Wilck, MD; Guang-Qian Xiao, MD; Alice C. Levine, MD; William B. Inabnet III, MD, FACS Submitted for publication January 24, 2014 Accepted for publication May 25, 2014 From The Mount Sinai Adrenal Center, Icahn School of Medicine at Mount Sinai, New York, New York. *Co-first authors Address correspondence to Dr. Sandi-Jo Galati, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place #1055, New York, NY 10029. Email: [email protected]. Published as a Rapid Electronic Article in Press at http://www.endocrine practice.org on October 8, 2014. DOI: 10.4158/EP14036.RA To purchase reprints of this article, please visit: www.aace.com/reprints. Copyright © 2015 AACE. ABSTRACT Objective: Pheochromocytomas are complex tumors that require a comprehensive and systematic management plan orchestrated by a multidisciplinary team. Methods: To achieve these ends, The Mount Sinai Adrenal Center hosted an interdisciplinary retreat where experts in adrenal disorders assembled with the aim of developing a clinical pathway for the management of pheochromocytomas. Results: The result was a consensus for the diagnosis, perioperative management, and postoperative management of pheochromocytomas, with specific recommendations from our team of adrenal experts, as well as a review of the current literature. Conclusion: Our clinical pathway can be applied by other institutions directly or may serve as a guide for institution-specific management. (Endocr Pract. 2015;21: 368-382) Abbreviations: CCB = calcium-channel blocker; CT = computed tomography; MEN = multiple endocrine neoplasia; MIBG = 123 I-metaiodobenzylguanidine; MRI = mag- netic resonance imaging; NF1 = neurofibromatosis type 1; PET = positron emission tomography; SPECT = single-photon emission computed tomography; VHL = von Hippel-Lindau INTRODUCTION Pheochromocytoma is a catecholamine-secreting tumor of the adrenal medulla that has an incidence of 2 to 8 cases per million annually (1,2). Given the rarity of pheo- chromocytomas, as well as the challenges of treatment, interdisciplinary management with a well-defined clinical pathway is essential. METHODS The Mount Sinai Adrenal Center convened an inter- disciplinary pheochromocytoma clinical pathway retreat that brought specialists together with expertise in adrenal disorders in order to delineate the optimal clinical pathway for the management of pheochromocytomas. The follow- ing disciplines participated in this 1-day retreat: anesthesia, cardiology, endocrinology, endocrine surgery, genetics, pathology, pharmacology, nuclear medicine, and radiol- ogy. The agenda was organized into 3 broad categories: diagnosis, perioperative management, and postoperative management and surveillance. RESULTS Diagnosis Biochemical Testing Pheochromocytomas arise from the chromaffin cells of the adrenal medulla, whereas paragangliomas (extra- adrenal pheochromocytomas) arise from sympathetic gan- glia. Norepinephrine is the predominant catecholamine synthesized by the sympathetic ganglia. Epinephrine is synthesized in the adrenal medulla by N-methylation of norepinephrine, catalyzed by the enzyme phenylethanol- amine N-methyltransferase, which is restricted to the chro- maffin cells of the medulla and induced by cortisol from the cortex (Fig. 1). Catecholamines continually leak from secretory granules and are inactivated by the enzyme cat- echol-O-methyltransferase into free normetanephrine and

Sandi-Jo Galati, MD*; Meena Said, MD

Embed Size (px)

Citation preview

Page 1: Sandi-Jo Galati, MD*; Meena Said, MD

368

Review Article

Sandi-Jo Galati, MD*; Meena Said, MD*; Rebekah Gospin, MD; Nikolina Babic, PhD;Karen Brown, MS; Eliza B. Geer, MD; Lale Kostakoglu, MD, MPH; Lawrence R. Krakoff, MD;Andrew B. Leibowitz, MD; Lakshmi Mehta, MD, FACMG; Simone Muller, BS, PharmD, BCPS;

Randall P. Owen, MD, MS, FACS; David S. Pertsemlidis, MD, FACS; Eric Wilck, MD;Guang-Qian Xiao, MD; Alice C. Levine, MD; William B. Inabnet III, MD, FACS

Submitted for publication January 24, 2014Accepted for publication May 25, 2014From The Mount Sinai Adrenal Center, Icahn School of Medicine at Mount Sinai, New York, New York. *Co-first authorsAddress correspondence to Dr. Sandi-Jo Galati, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place #1055, New York, NY 10029. Email: [email protected] as a Rapid Electronic Article in Press at http://www.endocrinepractice.org on October 8, 2014. DOI: 10.4158/EP14036.RATo purchase reprints of this article, please visit: www.aace.com/reprints.Copyright © 2015 AACE.

ABSTRACT

Objective: Pheochromocytomas are complex tumors that require a comprehensive and systematic management plan orchestrated by a multidisciplinary team. Methods: To achieve these ends, The Mount Sinai Adrenal Center hosted an interdisciplinary retreat where experts in adrenal disorders assembled with the aim of developing a clinical pathway for the management of pheochromocytomas. Results: The result was a consensus for the diagnosis, perioperative management, and postoperative management of pheochromocytomas, with specific recommendations from our team of adrenal experts, as well as a review of the current literature. Conclusion: Our clinical pathway can be applied by other institutions directly or may serve as a guide for institution-specific management. (Endocr Pract. 2015;21: 368-382)

Abbreviations:CCB = calcium-channel blocker; CT = computed tomography; MEN = multiple endocrine neoplasia; MIBG = 123I-metaiodobenzylguanidine; MRI = mag-netic resonance imaging; NF1 = neurofibromatosis type 1; PET = positron emission tomography; SPECT = single-photon emission computed tomography; VHL = von Hippel-Lindau

INTRODUCTION

Pheochromocytoma is a catecholamine-secreting tumor of the adrenal medulla that has an incidence of 2 to 8 cases per million annually (1,2). Given the rarity of pheo-chromocytomas, as well as the challenges of treatment, interdisciplinary management with a well-defined clinical pathway is essential.

METHODS

The Mount Sinai Adrenal Center convened an inter-disciplinary pheochromocytoma clinical pathway retreat that brought specialists together with expertise in adrenal disorders in order to delineate the optimal clinical pathway for the management of pheochromocytomas. The follow-ing disciplines participated in this 1-day retreat: anesthesia, cardiology, endocrinology, endocrine surgery, genetics, pathology, pharmacology, nuclear medicine, and radiol-ogy. The agenda was organized into 3 broad categories: diagnosis, perioperative management, and postoperative management and surveillance.

RESULTS

Diagnosis

Biochemical Testing Pheochromocytomas arise from the chromaffin cells of the adrenal medulla, whereas paragangliomas (extra-adrenal pheochromocytomas) arise from sympathetic gan-glia. Norepinephrine is the predominant catecholamine synthesized by the sympathetic ganglia. Epinephrine is synthesized in the adrenal medulla by N-methylation of norepinephrine, catalyzed by the enzyme phenylethanol-amine N-methyltransferase, which is restricted to the chro-maffin cells of the medulla and induced by cortisol from the cortex (Fig. 1). Catecholamines continually leak from secretory granules and are inactivated by the enzyme cat-echol-O-methyltransferase into free normetanephrine and

Page 2: Sandi-Jo Galati, MD*; Meena Said, MD

369

metanephrine (3). Free normetanephrine and metaneph-rine circulate in the plasma in low concentrations and have short half-lives, undergoing further sulfate conjugation by sulfotransferase isoenzyme (3,4) (Fig. 1). In contrast to the free metabolites, sulfated metanephrines are present in 20- to 40-fold higher concentrations, have a longer half-life, and are eliminated by urinary excretion (4). Catecholamine production by pheochromocytomas is variable and depends on the activity of the enzymes required for catecholamine synthesis (Fig. 1). This vari-ability in synthesis governs the clinical presentation of pheochromocytomas, which ranges from asymptomatic to mild, continuous symptoms to episodic pronounced symp-toms or crises (5,6). Up to 20% of pheochromocytomas are biochemically “silent,” with normal concentrations of plasma and urine catecholamines (6). However, these silent tumors metabolize catecholamines into metaneph-rines in increased concentrations in the plasma and urine and may only demonstrate elevated catecholamine concen-trations in the setting of a pheochromocytoma crisis (7). This provides a rationale for the measurement of catechol-amine metabolites as more sensitive and specific markers of excess over direct catecholamine measurement (8,9).

Plasma Versus Urinary Metanephrines The principle screening tests for diagnosis of pheo-chromocytoma are plasma free and urine fractionated metanephrines. The term “fractionated” historically has been used to distinguish the more recent methods that quantify both metanephrine and normetanephrine from the older chromatographic methods that measured only total metanephrines. Today, both plasma and urine screens report metanephrine and normetanephrine levels. Major reference laboratories use liquid chromatography-tandem mass spectrometry for measurement of catecholamine metabolites in plasma and urine. This is the most sensitive and specific methodology available and is free from ana-lytical drug interferences. Measurement of plasma free metanephrines is the recommended initial approach in screening for pheochro-mocytoma (10). This test is 96 to 100% sensitive and 89 to 98% specific for disease detection (4,11,12), depending upon the analytical method and patient population studied (11). Plasma metanephrines are elevated 4-fold over the upper limit of normal in 80% of patients with pheochromo-cytoma and are diagnostic of the disease (13). Clearance of plasma free metanephrines is independent of renal func-tion and is unaffected in patients with renal disease (9). Measurement of urine 24-hour fractionated metaneph-rines confirms mild to moderate elevations of plasma free metanephrine levels and is less sensitive (86 to 97%) and slightly less specific (86 to 95%) (11) (Fig. 2). Pre-analytical factors may affect measurement of both plasma and urine metanephrines. False elevations in plasma normetanephrine levels can be seen in patients over

age 60 years (14), whereas metanephrine levels are less affected (4). Plasma free normetanephrines and metaneph-rines are 30% and 12% higher, respectively, in the seated versus supine position (15). Diets rich in catecholamine-containing foods result in appreciable increases in urinary normetanephrines, but plasma normetanephrines are unaf-fected (15) (Table 1). False positives may also occur with use of certain medications, the most common of which are tricyclic antidepressants and phenoxybenzamine (13), due to elevation in blood catecholamine concentrations (Table 1). In equivocal cases, the clonidine suppression test can be used to distinguish true- from false-positive norepineph-rine elevations. Clonidine activates α-2 adrenergic recep-tors, inhibiting norepinephrine discharge. Persistent nor-epinephrine or normetanephrine elevations following the administration of clonidine suggests the presence of pheo-chromocytoma (16), whereas normalization of values (17) or a 50% decrease in concentration (18,19) has been used to define nonpathologic sympathetic activation. Eisenhofer et al (13) compared the diagnostic accuracy of norepineph-rine or normetanephrine concentrations following cloni-dine administration, reporting a specificity of 98 to 100% when postclonidine values remained above the upper limit of normal. However, the sensitivity of normetanephrine (96%) significantly exceeded norepinephrine (67%), con-firming 40% more cases (13).

Mount Sinai Recommendations Our approach is to measure plasma free metanephrines as the initial biochemical test given the ease of collection and high sensitivity, such that a negative value excludes pheochromocytoma. Values 3- to 4-times above the upper limit of normal are considered diagnostic of pheochro-mocytoma. Significant metanephrine elevations imply epinephrine excess, which localizes tumors to the adrenal medulla, whereas lone normetanephrine elevations suggest extra-adrenal disease. Mild elevations (false positives), particularly in plasma free normetanephrines, are common and require subsequent evaluation with 24-hour urine fractionated metanephrines. Patients are asked to avoid consumption of nonessential medications 1 week prior to testing. Chronic, essential medications such as tricyclic antidepressants, phenoxybenzamine, levodopa, α-methyldopa, and labet-alol should not be stopped abruptly and may require moni-tored titration to an alternative agent prior to biochemical testing.

Imaging Imaging plays a critical role in distinguishing pheo-chromocytomas from adrenal cortical tumors. Commonly used imaging modalities include computed tomography (CT), magnetic resonance imaging (MRI), and functional nuclear-medicine images.

Page 3: Sandi-Jo Galati, MD*; Meena Said, MD

370

Cortex    

   Medulla  

CH2—CH—C—O-­‐      

NH3  

O  

+  

HO  

CH2—CH—C—O-­‐      

NH3  

O  

+  

HO  OH  

CH2—CH2—NH2      

HO  OH  

CH—CH2—NH2      

HO  OH  

OH  

CH—CH2—NH—CH3      

HO  OH  

OH  

Tyrosine  

DOPA  

Dopamine  

Norepinephrine  

Epinephrine  

Tyrosine    Hydroxylase  

DOPA  Decarboxylase  

Dopamine b-­‐hydroxylase  

Phenylethanolamine  N-­‐transferase  

Cor�sol  

Epinephrine  

a)  

Norepinephrine  

b)  

Catechol-­‐O-­‐methyltransferase  

Metanephrine   Normetanephrine  cytoplasm  

Sulfotransferase    Isoenzyme    

Metanephrine  Sulfate    

Normetanephrine  Sulfate  

plasma  

Renal  Excre�on  

Fig. 1. Catecholamine synthesis and metabolism. (A) The catecholamine synthetic cascade begins in the adrenal medulla with tyrosine, which is converted to dopa in a rate-limiting reaction by tyrosine hydroxylase. Dopa is subsequently decarboxylated to dopamine, which is hydroxylated to norepinephrine. Epinephrine is synthesized from norepinephrine by the enzyme phenylethanolamine N-methyltransferase, which is restricted to the chromaffi n cells of the adrenal medulla and induced by cortisol from the adrenal cortex. (B) Norepinephrine and epinephrine continuously leak from their storage granules into the cell cytoplasm, where catechol-O-methyltransferase metabolizes norepinephrine and epinephrine into normetanephrine and metanephrine, respectively. Free normetanephrine and metanephrine circulate in the plasma and undergo further sulfate conjugation by sulfotransferase isoenzyme and are eliminated by urinary excretion.

On unenhanced CT scans, the attenuation of pheo-chromocytomas ranges from low density to that of soft tissue. Almost all have attenuation values greater than 10 Hounsfi eld units (HU) (20). On contrast-enhanced CT, pheochromocytomas demonstrate enhancement with delayed washout (21). This feature helps to distinguish pheochromocytomas from lipid-rich adenomas, which are characterized by low density on unenhanced CT scans (<10 HU) and early washout (21). Necrosis, cystic changes, and internal calcifi cations can also be seen in pheochro-mocytomas (21). The sensitivity of CT scans for detecting pheochromocytomas ranges from 90 to 100%, whereas the specifi city ranges from 70 to 80% (22). Pheochromocytoma sizes range from 1.2 to 15 cm, with a mean of 5.5 cm (Fig. 3) (20,23).

The typical appearance of a pheochromocytoma on MRI is hyperintense on T2-weighted images. However, internal hemorrhage and cystic components contribute to the heterogeneity of these lesions, and thus, 35% may not exhibit this hyperintense pattern on T2 imaging (24). On T1-weighted images, pheochromocytomas are often isoin-tense to muscle and hypointense to liver (20). The sensi-tivity and specifi city of MRI for detecting pheochromocy-toma is similar to that of CT (22). CT and MRI are very sensitive in detecting adre-nal pheochromocytomas but exhibit lower sensitivities for extra-adrenal and metastatic disease (25). Functional imaging using various radiotracers allows for whole-body images and detection of lesions outside the primary tumor site. The main modality of functional imaging used is the

Page 4: Sandi-Jo Galati, MD*; Meena Said, MD

371

Figure 2: Biochemical testing algorithm for diagnosing catecholamine excess.

INDICATIONS FOR TESTING:

• Hypertension • Tachycardia • Adrenal abnormality • Diaphoresis • Family history of MEN2 • von Hippel-Landau syndrome, • Familial paraganglioma syndrome • Neurofibromatosis type 1

ORDER Metanephrines, Plasma (free)

ORDER Metanephrines, fractionated by HPLC-MS/MS, Urine

Pheochromocytoma likely. Proceed with imaging studies

Rule out Pheochromocytoma

Not elevated

Mildly elevated or Intermediate

Not elevated

Elevated

High clinical suspicion for pheochromocytoma exists

Mildly elevated or

Intermediate

Repeat testing in 6-12 months

Yes

No No further testing

required

No

Yes High clinical suspicion for

pheochromocytoma

>4x upper limit of normal

Fig. 2. Biochemical testing algorithm for diagnosing catecholamine excess. HPLC-MS/MS = high-performance liquid chromatography-tandem mass spectrometry; MEN = multiple endocrine neoplasia.

Page 5: Sandi-Jo Galati, MD*; Meena Said, MD

372

Table 1Factors That May Affect Biochemical Testing for Pheochromocytoma, Causing False-Positive or False-Negative Results

Factor Examples

Effect on plasma free metanephrines

NMN MN 3MT

Effect on urine fractionated

metanephrines

NMN MN 3MT

Diet (4,15)d Fruits, nuts, potatoes, tomatoes, beans, fermented foods, processed meat, coffee (caffeic acid) (4), cereals (4)a

↔ ↔ ↑ ↔ ↔ ↑↑

Medications (4,13)

Tricyclic antidepressants

Serotonin norepinephrine reuptake inhibitors (81)

Monoamine oxidase inhibitors

Pheonoxybenzamine

Selective α-adrenergic blockers (doxazosin, terazosin, prazosin)

β-Adrenergic receptor blockers (atenolol, metoprolol, propranolol, labetalolb)

Calcium-channel blockers

Levodopa

Sympathomimetics (pseudoephedrine, nicotine, amphetamines)

Acetaminophena

Buspironec

Abrupt discontinuation of clonidine or BZD (11)

↑ ↑

↔ ↑

↑ ↑ ↔ ↔

↑ ↑

↑ ↑

↑ ↑ ↑ ↑

Physiologic influences

ExercisePosition (sitting vs. supine for 20 minutes before blood draw) (15,82)Cigarette smoking (83)d

↑ ↑

↑ ↑

Comorbidities or other characteristics

CHF (14)Untreated OSA (11)Renal dysfunction (4,84)e

HTN (4)Alcohol excess (11)Age >60 (4,14)Gender (female) (4)

↑ ↑↑ ↑↔ ↔ ↑ ↑ ↓

Abbreviations: 3MT = 3-methoxytyramine; BZD = benzodiazepine; CHF = congestive heart failure; HTN = hypertension; MN = metanephrine; NMN = normetanephrine; OSA = obstructive sleep apnea.a Interferes directly with the assay and analysis of plasma free normetanephrines.b Interferes directly with the assay and analysis of urinary catecholamines and metanephrines, variable increases in plasma epinephrine.c Interferes directly with the assay and analysis of urinary metanephrines.d Increases epinephrine and norepinephrine levels.e Diet most dramatically effects plasma and urine deconjugated normetanephrines and 3MT. Plasma and urine deconjugated metanephrines is unchanged. Therefore, it is recommended that patients fast beginning at midnight to avoid interference with biochemical testing (15). Renal dysfunction results in increases in plasma and urine deconjugated normetanephrines and metanephrines as well as vanillylmandelic acid levels (84).

Page 6: Sandi-Jo Galati, MD*; Meena Said, MD

373

123I-metaiodobenzylguanidine (MIBG) scan. MIBG is a guanethidine analog that is taken up by cells in the sym-pathomedullary system (26). Functional imaging can be done by labeling MIBG with either 131I or 123I. However, 123I is preferred because of its lower radiation dose, lack of beta emission, and shorter half-life (27). Its sensitivity ranges from 77 to 90%, with a specificity of 95 to 100% (20). Tracer activity in which the uptake is greater than that of the liver and marked asymmetry of the adrenal glands is suspicious for pheochromocytoma (28). MIBG is particu-larly useful in assisting when MRI/CT results are equivo-cal. MIBG allows for whole body evaluation and can detect extra-adrenal tumors, metastatic disease, and recurrence. Single-photon emission computed tomography (SPECT) is the standard application for cross-sectional evaluation but has limited spatial resolution for small lesions (29,30). The combination of functional and ana-tomic imaging in the form of 123I MIBG SPECT/CT and interpretation of SPECT data with MRI have recently emerged as useful imaging methods for pheochromocyto-mas (28). 123I MIBG SPECT/CT has a sensitivity of 87.5% and a specificity of 93.8% (28). However, investigations combining the use of SPECT and MRI are ongoing and have not reached a routine clinical platform yet (Fig. 4) (28). Positron emission tomography (PET) imaging with fluorodeoxyglucose (FDG) has limited sensitivity in benign pheochromocytomas, but malignant lesions are

readily detected (29,31). More specific imaging is pos-sible with the catecholamine precursor 6-[18F]fluoro-L-dihydroxyphenylalanine (18F-DOPA) or the catechol-amine 18F-fluorodopamine. Small studies have confirmed the feasibility of 18F-DOPA as a tracer for staging pheo-chromocytomas (29,32-35). Detection is also possible with a somatostatin receptor ligand using the PET radio-tracer 68Ga-dotatate. Preliminary studies have shown that 68Ga-dotatate PET/CT is useful as a first-line agent for those at high risk for paragangliomas and metastatic dis-ease (36). However, most studies involving research radiotrac-ers included only those with a high clinical suspicion of pheochromocytoma or paraganglioma, and sensitivities and specificities for an unbiased population have yet to be elucidated.

Mount Sinai Recommendations We recommend MRI or CT with contrast as the initial localization study. In the event that the diagnosis and/or imaging is equivocal, 123I MIBG scan can be considered. If the 123I MIBG scan is negative and a clinical suspicion still exists, then an FDG-PET scan should be performed.

Pre-operative Management Prevention of intra-operative hypertensive crisis is the principal goal of pre-operative management. Close intra-operative monitoring by an anesthesiologist familiar with

Fig. 3. Appearance of pheochromocytoma on different phases of a computed tomogra-phy scan with intravenous contrast.

Page 7: Sandi-Jo Galati, MD*; Meena Said, MD

374

the management of pheochromocytoma may abrogate the need for pre-operative medical treatment (37); however, most institutions have established a pre-operative treat-ment plan addressing both blood pressure control and vol-ume expansion to minimize surgical risk (38,39). α-Adrenergic blocking agents lower blood pressure by antagonizing catecholamine-stimulated vasoconstric-tion. They are the agents of choice in controlling pre-operative blood pressure, although there are no published data on the dose, agent selection, duration of therapy, or therapeutic goals (40). Phenoxybenzamine is a nonselec-tive, irreversible α-antagonist that is the preferred ini-tial agent due to its long duration of action and irrevers-ibility. Its tolerability is limited by its side effects (39). Alternative agents include the selective α1-adrenergic antagonists, which are used in patients that cannot toler-ate phenoxybenzamine and in those who require long-term medical treatment (41). β-Adrenergic blockade is indicated for management of tachycardia or arrhythmias induced by catecholamine excess after an α-antagonist has been introduced. β1-Selective drugs are preferred, as nonselec-tive β-blockers may antagonize β2-vasodilator action (42) (Table 2). Calcium-channel blockers (CCBs) can be used in conjunction with α- and β-adrenergic blockers or used alone as the primary agent for blood pressure control (41).

β-Adrenergic-stimulated renin secretion is present in a subset of patients with pheochromocytoma, so agents tar-geting the renin-angiotensin system, such as angiotensin-converting enzyme inhibitors and angiotensin-receptor blockers may improve blood pressure control in these patients (43). The less noxious side effect profiles of these agents make them useful alternatives for patients who can-not tolerate the α-blockading agents (Table 2). Catecholamine-synthesis inhibitors can be added in patients with catecholamine burdens that are unresponsive to adrenergic and calcium-channel blockade. α-Methyl-l-tyrosine (metyrosine) inhibits tyrosine hydroxylase (5,40), the rate-limiting step of the catecholamine biosynthetic cascade (Fig. 1), reducing catecholamine stores after 3 days of use (44). Because there is incomplete depletion of catecholamine stores, α-adrenergic blockade must be used in conjunction (5). The side effects of metyrosine can be disabling, including extrapyramidal symptoms, depres-sion, galactorrhea, and sedation, particularly with long-term use, and may require discontinuation (5). Therefore, its use should be reserved for widespread disease with intractable symptoms despite α-adrenergic blockade (45). Chronic vasoconstriction from catecholamine excess results in a volume-depleted state. A rational approach to minimizing pre-operative hypovolemia is institution of a liberalized salt diet in addition to oral fluid loading for

Fig. 4. Transaxial images of a large left adrenal mass in a 28-year-old man: computed tomography (CT) image (A); 123I-metaiodobenzylguanidine (MIBG) single-photon emission computed tomography (SPECT)/CT image (B); T2 turbo spin echo magnetic resonance (MR) image (C); and 123I-MIBG SPECT/MR image using a T1 fast field echo sequence (D). Both high signal intensity on T2-weighted magnetic resonance imaging and high tracer uptake are consistent with pheochromocytoma. This lesion contains areas of cystic necrosis.

Page 8: Sandi-Jo Galati, MD*; Meena Said, MD

375

Tabl

e 2

Pre-

oper

ativ

e, In

tra-

oper

ativ

e, a

nd P

osto

pera

tive

Phar

mac

olog

ic M

anag

emen

t of P

heoc

hrom

ocyt

oma

Cla

ssD

rug

and

dosin

gC

linic

al u

se

Pre-

oper

ativ

em

anag

emen

t

α-A

dren

ergi

c bl

ocka

de

Cal

cium

-cha

nnel

blo

ckad

e

Cat

echo

lam

ine

synt

hesis

inhi

bitio

n

Oth

ers

Non

sele

ctiv

e α

-blo

ckad

e: p

heno

xybe

nzam

ine

10-2

0 m

g 2-

3 tim

es/d

ay (1

00

mg

max

dai

ly)

Sele

ctiv

e α

-blo

ckad

e: d

oxaz

osin

1 m

g da

ily, t

eraz

osin

1 m

g 2

times

/day

, pr

azos

in 1

mg

3 tim

es/d

ay (a

ll 20

mg

max

dai

ly)

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

-----

-

Nife

dipi

ne S

R 3

0-12

0 m

g da

ilyN

icar

dipi

ne 3

0 m

g 2

times

/day

(120

mg

max

dai

ly)

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

-----

-

Met

yros

ine

250

mg

3-4

times

/day

(4 g

max

dai

ly)

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

-----

-β-

bloc

kade

, ang

iote

nsin

-con

vert

ing

enzy

me

inhi

bito

rs,

angi

oten

sin r

ecep

tor b

lock

ers

• Ph

enox

yben

zam

ine

is th

e pr

efer

red

agen

t as i

t pro

vide

s lon

g-ac

ting

nons

elec

tive

α-b

lock

ade

• Si

de e

ffect

s: e

xtre

me

fatig

ue, n

asal

con

gest

ion,

and

orth

osta

sis

• Si

de e

ffect

s are

less

pro

noun

ced

in se

lect

ive

α-b

lock

ade

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

• B

lock

s nor

epin

ephr

ine-

med

iate

d ca

lciu

m tr

ansp

ort i

nto

the

vasc

ular

sm

ooth

mus

cle

• C

an b

e us

ed a

s an

adju

nctiv

e ag

ent t

o α

- and

β-b

lock

ade

or in

pat

ient

s who

ca

nnot

tole

rate

α-b

lock

ing

agen

ts--

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

-----

-•

Not

rout

inel

y us

ed p

re-o

pera

tivel

y un

less

for s

hort

cour

se d

ue to

seve

re

sym

ptom

s•

Mai

n sh

ort-t

erm

side

effe

ct is

som

nole

nce

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

• β-

Blo

ckad

e ca

n be

inst

itute

d af

ter α

-blo

ckad

e fo

r tac

hyca

rdia

• β1

-sel

ectiv

e bl

ocka

de is

pre

ferr

ed (m

etop

rolo

l, bi

sopr

olol

and

ate

nolo

l)

Intr

a-op

erat

ive

man

agem

ent

Adr

ener

gic

bloc

kade

Cal

cium

-cha

nnel

blo

ckad

e

Vaso

dila

tors

Ane

sthe

tic a

gent

s/ ne

urom

uscu

lar

bloc

kade

Ana

lges

ia

Phen

tola

min

e 1-

5 m

g in

trave

nous

bol

uses

or i

nfus

ion

Esm

olol

0.5

mg/

kg o

ver 1

min

then

0.0

5 m

g/kg

/min

infu

sion

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

-----

Nic

ardi

pine

5 m

g/h

infu

sion

titra

tabl

e to

15

mg/

hC

levi

dipi

ne 1

-2 m

g/h

infu

sion

titra

tabl

e to

32

mg/

h--

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

---N

itrop

russ

ide

2 m

g/kg

/min

, not

to e

xcee

d 80

0 m

g/h

Mag

nesiu

m su

lfate

bol

us 4

0 m

g/kg

, inf

usio

n 1-

2 g/

h--

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

---Pr

opof

ol 4

0 m

g ev

ery

10 s,

not

to e

xcee

d 24

0 m

g

Etom

idat

e 0.

2-0.

6 m

g/kg

ove

r 30-

60 s

Thio

pent

al 3

-4 m

g/kg

Sevo

flura

ne 0

.5-3

% w

ith o

r with

out n

itrou

s oxi

deIs

oflur

ane

1-2%

with

nitr

ous o

xide

, add

0.5

-1%

with

oxy

gen

Des

flura

ne 2

.5-8

.5%

with

or w

ithou

t nitr

ous o

xide

Atr

acur

ium

0.4

-0.5

mg/

kg b

olus

, 0.0

8-0.

1 m

g/kg

20-

45 m

in a

fter b

olus

, rep

eat

mai

nten

ance

dos

e ev

ery

15-2

5 m

in O

R 0

.005

-0.0

1 m

g/kg

/min

infu

sion

for

mai

nten

ance

R

ocur

oniu

m b

olus

0.4

5-0.

6 m

g/kg

, 0.1

-0.2

mg/

kg e

very

20-

30 m

in fo

r m

aint

enan

ce O

R 0

.01-

0.01

2 m

g/kg

/min

infu

sion

for m

aint

enan

ce

Vecu

roni

um b

olus

0.0

8-0.

1 m

g/kg

, 0.0

1-0.

015

mg/

kg e

very

20-

45 m

in fo

r m

aint

enan

ce O

R

for c

ontin

uous

infu

sion

: int

ubat

ing

dose

of 0

.000

8-0.

001

mg/

kg fo

llow

ed 2

0-40

m

in la

ter w

ith 0

.000

8-0.

0012

mg/

kg/m

in in

fusi

on fo

r mai

nten

ance

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

---

Fent

anyl

1-3

mg/

kg/h

• Ph

ento

lam

ine

is a

non

sele

ctiv

e α

-blo

ckin

g ag

ent

• Es

mol

ol is

a β

-blo

cker

that

shou

ld b

e us

ed fo

r tac

hyar

rhyt

hmia

s--

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

-----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

• Th

iocy

anat

e le

vels

mus

t be

mea

sure

d w

ith n

itrop

russ

ide

use

• M

agne

sium

sulfa

te is

safe

in p

regn

ancy

and

chi

ldre

n--

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

-----

-

• D

esflu

rane

cau

ses s

ympa

thet

ic a

ctiv

atio

n

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

-*

O

ther

age

nts c

an b

e us

ed, f

enta

nyl i

s our

inst

itutio

nal p

refe

renc

e

Post

oper

ativ

em

anag

emen

t

Adr

ener

gic

bloc

kade

Cat

echo

lam

ine

synt

hesis

inhi

bitio

n

Phen

oxyb

enza

min

e 10

-20

mg

2-3

times

/day

, dox

azos

in 1

mg

daily

, ter

azos

in 1

m

g 2

times

/day

, pra

zosin

1 m

g 3

times

/day

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

Met

yros

ine

250

mg

3-4

times

/day

• α

-Blo

cker

s sho

uld

be re

sum

ed p

osto

pera

tivel

y fo

r res

idua

l or m

etas

tatic

di

seas

e--

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

----

---

• Po

stop

erat

ive

use

of m

etyr

osin

e is

indi

cate

d fo

r res

idua

l or m

etas

tatic

di

seas

e•

Side

effe

cts w

ith lo

ng-te

rm u

se in

clud

e di

arrh

ea, a

nxie

ty, n

ight

mar

es,

crys

tallu

ria, g

alac

torr

hea,

ext

rapy

rimid

al sy

mpt

oms i

n ad

ditio

n to

seda

tion

Page 9: Sandi-Jo Galati, MD*; Meena Said, MD

376

volume expansion once blood pressure control has been achieved, although limited data exist for this practice (39). There are no data to support inpatient admission for intra-venous fluid prior to surgery, although in patients with hemodynamic volatility or cardiomyopathy, this may be considered. Hyperglycemia due to catecholamine excess is a common metabolic derangement seen in patients with pheochromocytomas (46). The etiology is multifacto-rial, including increased gluconeogenesis and glycoge-nolysis by stimulation of hepatic β-adrenergic receptors (47), decreased insulin secretion by stimulation of pan-creatic α-adrenergic receptors (48), and decreased skel-etal muscle glucose uptake (49). All patients should be assessed pre-operatively for the presence of abnormal glucose metabolism with a fasting blood glucose mea-surement, an oral glucose tolerance test, or a hemoglobin A1c measurement.

Mount Sinai Recommendations The initial agent of choice in pre-operative blood pres-sure management is phenoxybenzamine due to its non-selective, irreversible action at the level of the receptor and long duration of action. Patients who cannot tolerate phenoxybenzamine should be transitioned to a selective α-blocker if tolerated or a CCB. Pre-operative α-blockade should be instituted a minimum of 7 days prior to surgery, unless the patient is normotensive. In cases of silent pheo-chromocytomas in which there is no α-blockade, consulta-tion with anesthesia prior to surgery is especially essential. Once the blood pressure is controlled, all patients are encouraged to increase salt and water intake for pre-oper-ative volume expansion. We do not suggest admission the day prior to surgery for intravenous fluid loading unless severe hemodynamic fluctuations or cardiomyopathy is present. All patients should undergo hemoglobin A1c and fast-ing glucose measurements at the time of diagnosis, given the increased incidence of hyperglycemia.

Perioperative Management

Operative Approach Adrenalectomy is the mainstay of management for pheochromocytomas. Laparoscopic surgery is the standard operative approach for the resection of pheochromocyto-mas. This can be done via a transabdominal or retroperito-neal route, depending on the size of the tumor. The superi-ority of the laparoscopic approach over the open approach has been established by numerous studies (50,51). When compared with open adrenalectomies, the laparoscopic method has lower mean operative time, shorter hospital stay, decreased need for intensive care, less blood loss, and lower pain medication requirement. The complication rates

are equivalent between the two methods. Tumor size does not affect outcome (50). Tumors larger than 9 cm can be removed safely with this method (50). The laparoscopic retroperitoneal approach has been shown to be a safe alternative to the laparoscopic transabdominal method for smaller adrenal tumors (52). An open approach is warranted when malignancy is suspected. Imaging characteristics raising suspicion for malignancy include large size, necrosis, hemorrhage, retained contrast on CT studies, and evidence of invasion into surrounding structures (53,54).

Mount Sinai Recommendations We recommend laparoscopic adrenalectomy in patients with pheochromocytomas, which can be per-formed safely with either the transabdominal laparoscopic approach or the retroperitoneal laparoscopic method. In the event that malignancy is suspected, open adrenalectomy is recommended.

Anesthesia The intra-operative management of patients with pheochromocytoma is complex and challenging. Life-threatening fluctuations in blood pressure and cardiovascu-lar collapse can occur. Several measures are recommended in order to mitigate the chance of cardiovascular complica-tions. Euvolemia should be maintained by administering a balanced salt solution (e.g., lactated Ringers, Plasmalyte A) at a 10 to 30 mL/kg/hour rate based upon blood loss and hemodynamic assessment. Induction agents that have been safely used include propofol, etomidate, and thiopental (37,38,55). Etomidate may provide more cardiovascular stability, but myoc-lonus, increased postoperative nausea/vomiting, and decreased steroid-genesis need to be considered. Agents that cause catecholamine release, such as ketamine and ephedrine (37,56,57), must be avoided, as the response is unpredictable. Muscle relaxants that are used with success include atracurium, rocuronium, and vecuronium (55,58,59). Pancuronium is not recommended given its vagolytic effect and prolonged duration of action (58,60). Maintenance of anesthesia with any volatile agent is safe (57,58,61,62), and even though desflurane causes sympathetic activation, it too has been used in these cases (58,63). Mount Sinai Recommendations We recommend the use of propofol as an induction agent and sevoflurane or desflurane as maintenance drugs. We recommend maintaining euvolemia by administering a balanced salt solution. Vecuronium and rocuronium should be used for muscle relaxation, whereas fentanyl should be the mainstay of analgesia.

Page 10: Sandi-Jo Galati, MD*; Meena Said, MD

377

Intra-operative Managementof Hypertension

Routine use of an arterial line is recommended for intra-operative blood pressure monitoring and should be placed before the induction of anesthesia. A central line is part of routine care for many teams but may be used more selectively. Intra-operative hypertension can be managed with a variety of agents. In a review of the current data, the most commonly used agents were nitrates (67%) and β-adrenergic blockers (47%). α-Adrenergic blockers were used in 33% of cases. In the control of intra-operative hypertension, 1 to 3 different agents are often used (64). Sodium nitroprusside is used as a first-line agent in treating intra-operative hypertension. Its onset of action is immediate and recovery occurs within 1 to 2 minutes. Toxic metabolites are a concern when it is administered in high doses or for greater than 4 to 72 hours (58). Nitroglycerin is also used to control intra-operative hypertension. Its onset of action and duration are rapid. Phentolamine is an α-adrenergic blocker that is used commonly for intra-operative control of hypertension. Its action is immediate, lasting 15 to 30 minutes, and it can be used as a continuous infusion (65). Esmolol is a β-adrenergic blocker used for blood pres-sure and heart rate control intra-operatively. It is particu-larly helpful when patients exhibit tachycardia. Its onset of action is 60 seconds and lasts 10 to 20 minutes (58). Magnesium sulfate is also used for intra-operative hypertension management. It is safe and effective to use in children and pregnant patients (66). Clevidipine butyrate is a novel short-acting CCB. Its onset is approximately 2 to 4 minutes and lasts approxi-mately 5 to 15 minutes. Its quick onset and rapidly titrat-able qualities make it a desirable agent for intra-operative hypertension (66). Another benefit is its cost. Clevidipine costs approximately $130.00 per case. In contrast, phentol-amine costs approximately $9,000.00 per case. Nicardipine is a second-generation CCB commonly used for intra-operative hypertension. Its half-life is 3 to 7 minutes and it decreases blood pressure in a dose-depen-dent fashion, with a response time of 1 to 3 minutes (67).

Mount Sinai Recommendations We recommend the use of clevidipine, phentolamine, and esmolol as first-line agents for intra-operative control of hypertension. Clevidipine is preferred, as it is more cost effective. Second-line agents include nicardipine and sodium nitroprusside.

Postoperative Managementand Surveillance

Immediate Postoperative Period Close inpatient observation is required for 24 hours after surgery because withdrawal of catecholamine excess

may result in significant hemodynamic changes (37,68). Postoperative hypotension results from a number of dif-ferent factors, including the loss of peripheral vasocon-striction after tumor removal and the prolonged effects of irreversible α-adrenergic blockade. This may result in catecholamine-resistant hypotension in which vasopressin is required for hemodynamic support (37,69). Postoperative hypoglycemia may also occur as a result of transient catecholamine deficiency and hyper-insulinemia. Hypoglycemia is worsened by prolonged α-adrenergic blockade. Thus, close monitoring of blood glucose is needed in the postoperative period (37,70-72).

Mount Sinai Recommendations We recommend close hemodynamic monitoring of all patients in the 24 hours following surgery. This can be done in the typical floor-bed setting. Monitoring includes close attention to vitals, urine output, and fluid status. The use of a monitored step-down unit is not routinely neces-sary. However, if there is pre-operative or intra-operative concern about hemodynamic instability, then a step-down unit would be appropriate. We recommend blood glucose monitoring with fingerstick or serum glucose every 4 to 6 hours for 24 hours postoperatively.

Biochemical Monitoring andResidual Hypertension

Plasma free or urinary deconjugated metanephrines should be tested 2 to 6 weeks postoperatively to assess for complete tumor resection (10,38). Normalization of biochemical parameters does not unequivocally indicate cure due to the potential presence of residual microscopic disease. Therefore, biochemical screening should be per-formed annually (10). Hypertension typically resolves within days of sur-gery; however, residual hypertension may persist up to 2 months postoperatively. In the setting of normalization of plasma or urinary metanephrines after tumor resection, residual hypertension present after 2 months is typically essential hypertension and should be managed following the seventh report of the Joint National Committee (JNC 7) hypertension guidelines (73). Persistent metanephrine elevations may signal resid-ual tumor, multifocal disease, or metastatic disease and determine the need for additional imaging. Hypertension and symptoms of catecholamine excess in patients with residual or metastatic disease should be managed as they were pre-operatively, primarily with α-adrenergic block-ade; however, the selective α-blocking agents (prazosin, doxazosin, or terazosin) may be employed in these cases to minimize the side effects of expected long-term use. In patients with refractory disease, metyrosine can be used to manage the catecholamine burden (5,40). Because of its side effects, particularly with long-term use, mety-rosine should be reserved for patients with widespread

Page 11: Sandi-Jo Galati, MD*; Meena Said, MD

378

disease and intractable symptoms despite α-adrenergic blockade (45).

Mount Sinai Recommendations Biochemical testing with plasma free metanephrines is performed during the initial outpatient follow-up visit 2 to 6 weeks postoperatively and annually thereafter. In patients in whom biochemical remission has been demon-strated, persistent hypertension is managed according to JNC-7 guidelines for essential hypertension.

Genetic Testing Historically, approximately 10% of pheochromocyto-mas and paragangliomas were estimated to be due to one of the known hereditary syndromes conferring susceptibility to the tumors: neurofibromatosis type 1(NF1 gene), multi-ple endocrine neoplasia (MEN) 2 (RET gene), von Hippel-Lindau (VHL) disease (VHL gene), and, rarely, MEN 1 (MEN1 gene) (74). In recent years, multiple studies have shown that this figure is higher, particularly with the discovery of addi-tional susceptibility genes. These include germline muta-tions in the genes encoding components of the succinate dehydrogenase complex (i.e., SDHB, SDHC, SDHD, SDHA, and SDHAF2), as well as mutations in TMEM127, MAX, KIF1Bβ, EGLN1/PHD2, and HIF2A (75). Germline mutations in RET, VHL, SDHD, and SDHB were identified in one-fourth of patients with nonsyn-dromic sporadic disease (76). In a subsequent retrospective study of 139 patients, 41% had mutations in one of five genes (SDHB, SDHD, VHL, RET, NF1) (77). In patients with at least one paraganglioma outside the adrenal gland, 53% had an identified mutation (77). Most familial cases of pheochromocytoma or paraganglioma and 10 to 20% of sporadic cases will have a germline mutation in one of the 10 known genes (78). Several clinical algorithms have been suggested to guide genetic testing, focusing on limiting the cost of test-ing by identifying the genes most likely to be involved in each clinical scenario based on the characteristics of the tumor (Table 3). A next-generation sequencing (NGS) strategy allow-ing simultaneous analysis of nine of the susceptibility genes (all but NF1) has been recently validated (79). This approach is likely to be more cost effective. An inevita-ble result of this technology is the detection of sequence variants of unknown significance, which require complex interpretation before any conclusions can be made regard-ing their pathogenicity. As experience grows with sequenc-ing these genes, information will be refined and fewer vari-ants of unknown significance will be reported. Risk factors for the presence of an underlying genetic syndrome or susceptibility include extra-adrenal pheochro-mocytoma, head or neck paraganglioma, age younger than 45 years, bilateral or multicentric disease, and malignant

tumors (80). Family history of pheochromocytoma or paraganglioma or clinical presentation consistent with one of the known hereditary syndromes should also prompt genetic counseling and testing (74).

Mount Sinai Recommendations Our practice is to refer all patients with pheochromo-cytoma or paraganglioma for genetic counseling and test-ing who have:

(1) syndromic features suggestive of NF1, VHL, or MEN2. Hallmark features include personal or family history of multiple café-au-lait spots, neurofibromas, renal cell carcinoma, heman-gioblastomas, medullary thyroid cancer, or hyperparathyroidism;

(2) a family history of pheochromocytomas or para gangliomas or sudden unexplained death of a family member at a young age;

(3) presence of extra-adrenal or multifocal tumors;(4) age of diagnosis prior to 50 years;(5) malignant tumors.

Because the estimate for germline mutations in spo-radic pheochromocytomas or paragangliomas approaches 10 to 20%, screening of all patients with pheochromocyto-mas or paragangliomas should be strongly considered. In the future, the availability of NGS panels will enable more widespread and comprehensive testing at lower cost.

DISCUSSION

This review presents our institutional approach in treating patients with pheochromocytomas and paragan-gliomas. Given the complexity of the disease and potential for serious adverse outcomes, a collaborative approach is required for the systematic management of pheochromocy-tomas and paragangliomas. This is achieved by following an evidence-based outline of specific therapeutic goals and measures for each time period (pre-operative diagnosis, perioperative management, and postoperative manage-ment and surveillance). In order to implement this strategy, a multidisciplinary team involving experts who are experi-enced in the evaluation and treatment of pheochromocyto-mas is required.

CONCLUSION

Our clinical pathway can be applied directly or used by others as a platform from which they can develop their institutional methods for the treatment and management of this neuroendocrine disease.

DISCLOSURE

The authors have no multiplicity of interest to disclose.

Page 12: Sandi-Jo Galati, MD*; Meena Said, MD

379

Tabl

e 3

Path

ophy

siolo

gy a

nd F

eatu

res o

f Kno

wn

Synd

rom

es a

nd S

usce

ptib

ilitie

s A

ssoc

iate

d W

ith P

heoc

hrom

ocyt

oma

and

Para

gang

liom

as (7

4,75

)

Gen

e/pa

thop

hysio

logy

Clin

ical

feat

ures

Syndromes

Mul

tiple

end

ocri

ne n

eopl

asia

(MEN

) 2a

and

2b:

activ

atin

g m

utat

ion

of th

e RE

T pr

oto-

onco

gene

• Ep

inep

hrin

e an

d no

repi

neph

rine

secr

etio

n, 5

0% b

ilate

ral,

typi

cally

ben

ign,

typi

cally

intra

-adr

enal

• M

EN 2

A: m

edul

lary

thyr

oid

canc

er, h

yper

para

thyr

oidi

sm

• M

EN 2

B: m

edul

lary

thyr

oid

canc

er, m

ucos

al g

angl

ione

urom

as, m

arfa

noid

feat

ures

von

Hip

pel-L

inda

u (V

HL)

dise

ase:

VH

L ge

ne

prom

otes

deg

rada

tion

of th

e hy

poxi

a-in

duci

ble

fact

or

(HIF

) 1α

• N

orep

inep

hrin

e se

cret

ion,

bila

tera

l, ty

pica

lly b

enig

n, ty

pica

lly in

tra-a

dren

al

• C

ereb

ella

r and

spin

al h

eman

giob

last

omas

, ret

inal

hem

angi

omas

, pan

crea

tic n

euro

endo

crin

e tu

mor

s, ep

idid

ymal

cy

sts,

rena

l cel

l car

cino

mas

Neu

rofib

rom

atos

is (N

F) ty

pe 1

: los

s-of

-fun

ctio

n m

utat

ion

of N

F1, a

tum

or su

ppre

ssor

• Ep

inep

hrin

e se

cret

ion,

uni

late

ral,

intra

-adr

enal

, ben

ign

• C

afé-

au-la

it sp

ots,

neur

ofibr

omas

, iris

ham

arto

mas

, ski

nfol

d fr

eckl

ing,

opt

ic n

erve

glio

mas

, phe

noid

bon

e dy

spla

sia

MEN

1: M

EN1

tum

or su

ppre

ssor

gen

e•

Rar

e ph

eoch

rom

ocyt

omas

• A

ssoc

iate

d w

ith p

ituita

ry tu

mor

s, hy

perp

arat

hyro

idis

m, a

nd p

ancr

eatic

tum

ors

Susceptibilities

Succ

inat

e de

hydr

ogen

ase

(SD

H) c

ompl

ex: o

xidi

zes

succ

inat

e in

to fu

mar

ate,

m

utat

ions

resu

lt in

su

ccin

ate

accu

mul

atio

n w

ith

stab

iliza

tion

of H

IF-1

α

SDH

B•

Nor

epin

ephr

ine

and

dopa

min

e, e

xtra

-adr

enal

, hig

h ra

te o

f mal

igna

ncy

• A

ssoc

iatio

ns: r

enal

cel

l car

cino

ma,

bre

ast c

ance

r, ga

stro

inte

stin

al st

rom

al tu

mor

s, pa

pilla

ry th

yroi

d ca

ncer

SDH

C•

Nor

epin

ephr

ine

and

dopa

min

e, e

xtra

-adr

enal

esp

ecia

lly h

ead

and

neck

, ben

ign

SDH

D•

Nor

epin

ephr

ine

and

dopa

min

e, e

xtra

-adr

enal

esp

ecia

lly h

ead

and

neck

, mul

tifoc

al, t

ypic

ally

ben

ign

SDH

A•

Typi

cally

ext

ra-a

dren

al, a

lso

asso

ciat

ed w

ith L

eigh

synd

rom

e (n

euro

dege

nera

tive

diso

rder

)SD

HAF

2•

Typi

cally

ext

ra-a

dren

al e

spec

ially

hea

d an

d ne

ck, m

ultif

ocal

Tran

smem

bran

e (T

MEM

) pro

tein

127

: TM

EM12

7 is

a tu

mor

supp

ress

or in

the

rapa

myc

in p

athw

ay•

Epin

ephr

ine

and

nore

pine

phrin

e se

cret

ion,

intra

-adr

enal

, ben

ign

MY

C-a

ssoc

iate

d fa

ctor

X: M

AX re

gula

tes c

ellu

lar

prol

ifera

tion

and

apop

tosi

s•

Epin

ephr

ine

and

nore

pine

phrin

e se

cret

ion,

intra

-adr

enal

, ofte

n bi

late

ral,

mal

igna

nt c

ases

repo

rted

Kin

esin

fam

ily m

embe

r 1B:

KIF

1B in

duce

s ap

opto

sis

• Si

ngle

pat

ient

with

phe

ochr

omoc

ytom

a, a

ssoc

iate

d w

ith n

euro

blas

tom

as

EGLN

1/PH

D2

enco

des P

HD

2 en

zym

e w

hich

is

invo

lved

in H

IF-1

α d

egra

datio

n•

Extra

-adr

enal

tum

ors a

nd e

ryth

rocy

tosi

s

Hyp

oxia

-indu

cibl

e fa

ctor

HIF

2A g

ene

• A

ssoc

iate

d w

ith p

olyc

ythe

mia

• A

lso

repo

rted

in so

mat

ic m

utat

ions

Page 13: Sandi-Jo Galati, MD*; Meena Said, MD

380

REFERENCES

1. Stenström G, Svärdsudd K. Pheochromocytoma in Sweden 1958-1981. An analysis of the national cancer reg-istry data. Acta Med Scand. 1986;220:225-232.

2. Sheps SG, Jian NS, Klee GG. Diagnostic evaluation of pheochromocytoma. Endocrinol Metab Clin North Am. 1988;17:397-414.

3. Schultz C, Eisenhofer G, Lehnert H. Principles of cat-echolamine biosynthesis, metabolism and release. Front Horm Res. 2004;31:1-25.

4. Eisenhofer G, Lenders JW, Pacek K. Biochemical diagnosis of pheochromocytoma. Front Horm Res. 2004;31:76-106.

5. Pacek K. Preoperative management of the pheochromocy-toma patient. J Clin Endocrinol Metab. 2007;92:4069-4079.

6. Bravo EL. Evolving concepts in the pathophysiology, diagnosis and treatment of pheochromocytoma. Endocr Rev. 1994;15:356-368.

7. Eisenhofer G, Keiser H, Friberg P, et al. Plasma meta-nephrines are markers of pheochromocytoma produced by catecol-O-methyltransferase within tumors. J Clin Endocrinol Metab. 1998;83:2175-2185.

8. Eisenhofer G, Tischler AS, de Krijger RR. Diagnostic tests and biomarkers for pheochromocytoma and extra-adrenal paraganglioma: from routine laboratory methods to disease stratification. Endocr Pathol. 2012;23:4-14.

9. Eisenhofer G. Screening for pheochromocytomas and paragangliomas. Curr Hypertens Rep. 2012;14:130-137.

10. Pacek K, Eisenofer G, Ahlman H, et al. Pheochromocytoma: recommendations for clinical practice from the First International Symposium. Nat Clin Pract Endocrinol Metab. 2007;3:92-102.

11. Schwartz, GL. Screening for adrenal-endocrine hyper-tension: overview of accuracy and cost-effectiveness. Endocrinol Metab Clin North Am. 2011;40:279-294.

12. Unger N, Pitt C, Schmidt IL, et al. Diagnostic value of various biochemical parameters for the diagnosis of pheochromocytoma in patients with adrenal mass. Eur J Endocrinol. 2006;154:409-417.

13. Eisenhofer G, Goldstein DS, Walther MM, et al. Biochemical diagnosis of pheochromocytoma: how to distinguish true-from false-positive test results. J Clin Endocrinol Metab. 2003;88:2656-2666.

14. Sawka, AM, Jaeschke R, Singh RJ, Young WF Jr. A comparison of biochemical tests for pheochromocytoma: measurement of fractionated metanephrines compared with combination of 24-hour urinary metanephrines and cate-cholamines. J Clin Endocrinol Metab. 2003;88:553-558.

15. de Jong WH, Eisenhofer G, Post WJ, Muskiet FA, de Vries EG, Kema IP. Dietary influences on plasma and urinary metanephrines: implications for diagnosis of cat-echolamine-producing tumors. J Clin Endocrinol Metab. 2009;94:2841-2849.

16. Bravo EL, Tarazi RC, Fouad FM, Vidt DG, Gifford RW Jr. Clonidine-suppression test: a useful aid in the diagnosis of pheochromocytoma. N Engl J Med. 1981;305:623-626.

17. Bravo EL, Gifford Jr RW. Current concepts. Pheochromocytoma: diagnosis, localization and manage-ment. N Engl J Med. 1984;311:1298-1303.

18. Grossman E, Goldstein DS, Hoffman A, Keiser HR. Glucagon and clonidine testing in the diagnosis of pheo-chromocytoma. Hypertension. 1991;17(6 Pt 1):733-741.

19. Elliott WJ, Murphy MB. Reduced specificity of the cloni-dine suppression test in patients with normal plasma cat-echolamine levels. Am J Med. 1988;84(3 Pt 1):419-243.

20. Leung K, Stamm M, Raja A, Low G. Pheochromocytoma: the range of appearances on ultrasound, CT, MRI, and func-tional imaging. AJR Am J Roentgenol. 2013;200:370-378.

21. Blake MA, Kalra MK, Maher MM. Pheochromocytoma: an imaging chameleon. Radiographics. 2004;24(suppl 1):S87-S99.

22. Ilias I, Pacak K. Current approaches and recommended algorithm for the diagnostic localization of pheochromocy-toma. J Clin Endocrinol Metab. 2004;89:479-491.

23. Caiafa RO, Izquierdo RS, Villalba LB, et al. Diagnosis and management of adrenal incidentaloma. Radiologia. 2011;53:516-530.

24. Varghese JC, Hahn PF, Papanicolaou N, Mayo-Smith WW, Gaa JA, Lee MJ. MR differentiation of pheochromo-cytoma from other adrenal lesions based on qualitative anal-ysis of T2 relaxation times. Clin Radiol.1997;52:603-606.

25. Lenders JW, Eisenhofer G, Mannelli M, Pacak K. Phaeochromocytoma. Lancet. 2005;366:665-675.

26. Intenzo CM, Jabbour S, Lin HC et al. Scintigraphic imaging of body neuroendocrine tumors. Radiographics. 2007;27:1355-1369.

27. Baez JC, Jagannathan JP, Krajewski K, et al. Pheochromocytoma and paraganglioma: imaging charac-teristics. Cancer Imaging. 2012;12:153-162.

28. Derlin T, Busch JD, Wisotzki et al. Intraindividual Comparison of 123I- mIBG SPECT/MRI, 123I- mIBG SPECT/CT, and MRI for the detection of adrenal pheo-chromocytoma in patients with elevated urine or plasma catecholamines. Clin Nucl Med. 2013;38:e1-e6.

29. Timmers HJ, Kozupa A, Chen CC, et al. Superiority of fluorodeoxyglucose positron emission tomography to other functional imaging techniques in the evaluation of meta-static SDHB-associated pheochromocytoma and paragan-glioma. J Clin Oncol. 2007;25:2262-2269.

30. Van der Haarst E, de Herder WW, Bruining HA, et al. [(123)I]metaiodobenzylguanidine and [(111)In]octreotide uptake in benign and malignant pheochromocytomas. J Clin Endocrinol Metab. 2001;86:685-693.

31. Ilias I, Yu J, Carrasquillo JA, et al. Superiority of 6-[18F]-fluorodopamine positron emission tomography versus [131I]-metaiodobenzylguanidine scintigraphy in the localization of metastatic pheochromocytoma. J Clin Endocrinol Metab. 2003;88:4083-4087.

32. Hoegerle S, Nitzsche E, Altehoefer C, et al. Pheochromocytomas: detection with 18F DOPA whole body PET—initial results. Radiology. 2002;222:507-512.

33. Taïeb D, Timmers HJ, Hindié E, et al. EANM 2012 guidelines for radionuclide imaging of phaeochro-mocytoma and paraganglioma. Eur J Nucl Med Mol Imaging. 2012;39:1977-1995.

34. Ilias I, Chen CC, Carrasquillo JA, et al. Comparison of 6-18F-fluorodopamine PET with 123I-metaiodo-benzylguanidine and 111in-pentetreotide scintigraphy in localization of nonmetastatic and metastatic pheochromo-cytoma. J Nucl Med. 2008;49:1613-1619.

35. Imani FImani F, Agopian VG, Auerbach MS, et al. 18F-FDOPA PET and PET/CT accurately localize pheo-chromocytomas. J Nucl Med. 2009;50:513-519.

36. Maurice JB, Troke R, Win Z, et al. A comparison of the performance of 68Ga-DOTATATE PET/CT and ¹²³I-MIBG SPECT in the diagnosis and follow-up of phaeochromocy-toma and paraganglioma. Eur J Nucl Med Mol Imaging. 2012;39:1266-1270

37. Lentschener C, Gaujoux S, Tesniere A, Dousset B. Point of controversy: perioperative care of patients undergoing pheochromocytoma removal—time for a reappraisal? Eur J Endocrinol. 2011;165:365-373.

Page 14: Sandi-Jo Galati, MD*; Meena Said, MD

381

38. Därr R, Lenders JW, Hofbauer LC, Naumann B, Bornstein SR, Eisenhofer G. Pheochromocytoma-update on disease management. Ther Adv Endocrinol Metab. 2012;3:11-26.

39. Young WF Jr. Endocrine hypertension. In: Melmed S, Polonsky KS, Larsen PR, Kronenberg HM, eds. Williams Textbook of Endocrinology. 12th ed. Elsevier; 2011.

40. Phitayakorn R, McHenry CR. Perioperative consid-erations in patients with adrenal tumors. J Surg Onc. 2012;106:604-610.

41. Bravo EL, Tagle R. Pheochromoocytoma: state-of-the-art and future prospects. Endocr Rev. 2003;24:539-553.

42. Prys-Roberts C. Phaeochromocytoma-recent progress in its management. Br J Anaesth. 2000;85:44-57.

43. Krakoff LR, Garbowit D. Adreno-medullary hyperten-sion: a review of syndromes, pathophysiology, diagnosis, and treatment. Clin Chem. 1991;37(10 Pt 2):1849-1853.

44. Brogden RN, Heel RC, Speight TM, Avery GS. alpha-Methyl-L-tyrosine: a review of its pharmacology and clini-cal use. Drugs. 1981;21:81-89.

45. Kopf D, Goretzki PE, Lehnert H. Clinical management of malignant adrenal tumors. J Cancer Res Clin Oncol. 2001;127:143-155.

46. Chrousos GP. Stressors, stress, and neuroendocrine inte-gration of the adaptive response. The 1997 Hans Selye Memorial Lecture. Ann N Y Acad Sci. 1998;851:311-335.

47. Bearn AG, Billing B, Sherlock S. The effect of adren-aline and noradrenaline on hepatic blood flow and splanchnic carbohydrate metabolism in man. J Physiol. 1951;115:430-441.

48. Isles CG, Johnson JK. Pheochromocytoma and dia-betes mellitus: Further evidence that alpha 2 receptors inhibit insulin release in man. Clin Endocrinol (Oxf). 1983;18:37-41.

49. Sacca L, Vigorito C, Cicala M, Ungaro B, Sherwin RS. Mechanisms of epinephrine-induced glucose intolerance in normal humans. J Clin Invest.1982;69:284-293.

50. Toniato A, Boschin IM, Opocher G, Guolo A, Pelizzo M, Mantero F. Is the laparoscopic adrenalectomy for pheochro-mocytoma the best treatment? Surgery. 2007;141:723-727.

51. Matsuda T, Murota T, Oguchi N, Kawa G, Muguruma K. Laparoscopic adrenalectomy for pheochromocytoma: a literature review. Biomed Pharmacother. 2002;56(suppl 1):132s-138s.

52. Dixon PV, Alex CG, Grubbs EG, et al. Posterior retroper-itoneoscopic adrenalectomy is a safe and effective alter-native to transabdominal laparoscopic adrenalectomy for pheochromocytoma. Surgery. 2011;150:452-458.

53. Szolar DH, Korobkin M, Reittner P, et al. Adrenocortical carcinomas and adrenal pheochromocytomas: mass and enhancement loss evaluation at delayed contrast-enhanced CT. Radiology. 2005;234:479-485.

54. Smith SM, Patel SK, Turner DA, Matalon TA. Magnetic resonance imaging of adrenal cortical carcinoma. Urol Radiol. 1989;11:1-6.

55. Hull CJ. Pheochromocytoma: diagnosis, preoperative preparation, and anaesthetic management. Br J Anaesth. 1986;58:1453-1468.

56. Desmonts JM, Marty J. Anaesthetic management of patients with pheochromocytoma. Br J Anaesth. 1984;56: 781-789.

57. Sumikawa K, Amakata Y. The pressor effect of droperi-dol on a patient with pheochromocytoma. Anesthesiology. 1977;46:359-361.

58. Kinney MA, Narr BJ, Warner MA. Perioperative man-agement of pheochromocytoma. J Cardiothorac Vas Anesth. 2002;16:359-369.

59. Brunjes S, Johns VJ Jr, Crane MG. Pheochromcytoma: postoperative shock blood volume. N Engl J Med. 1960; 262:393-396.

60. Lavery GG, Clarke RS, Watkins J. Histaminoid responses to atracurium, vecuronium, and tubocurarine. Ann Fr Aneth Reanim. 1985;4:180-183.

61. Kinney MA, Warner ME, VanHeerden JA, et al. Perianesthetic risks and outcomes of pheochromo-cytoma and paraganglioma resection. Anesth Analg. 2000;91:1118-1123.

62. Janeczko GF, Ivankovich AD, Glisson SN, Hayman HJ, El-Etr AA, Albrecht RF. Enflurane anesthesia for surgical removal of pheochromocytoma. Anesth Analg. 1977;56:62-67.

63. Lippman M, Ford M, Lee C, et al. Use of desflurane during resection of pheochromocytoma. Br J Anaesth. 1994;72:707-709.

64. Hariskov S, Schumann R. Intraoperative management of patients with incidental catecholamine producing tumors: a literature review and analysis. J Anaesthesiol Clin Pharmacol. 2013;29:41-46.

65. McMillian WD, Trombley BJ, Charash WE, Christian RC. Phentolamine continuous infusion in a patient with pheochromocytoma. Am J Health Syst Pharm. 2011:68:130-134.

66. Lord M, Augoustides JG. Perioperative management of pheochromocytoma: focus on magnesium, clevidipine, and vasopressin. J Cardiothorac Vasc Anesth. 2012;26:526-531.

67. Halpern NA, Goldberg M, Neely C, et al. Postoperative hypertension: a multicenter, prospective, randomized com-parison between intravenous nicardipine and sodium nitro-prusside. Crit Care Med. 1992;20:1637-1643.

68. Mannelli M. Management and treatment of pheochro-mocytomas and paragangliomas. Ann NY Acad Sci. 2006;1073:405-416.

69. Augoustides JG, Abrams M, Berkowitz D, Fraker D. Vasopressin for hemodynamic rescue in catecholamine resistant vasoplegic shock after resection of massive pheo-chromocytoma. Anesthesiology. 2004;101:1022-1024.

70. Meeke RI, O’Keeffe JD, Gaffney JD. Pheochromocytoma removal and postoperative hypoglycemia. Anaesthesia. 1985;40:1093-1096.

71. Isles CG, Johnson JK. Pheochromocytoma and diabetes mellitus: further evidence that alpha 2 receptors inhibit insu-lin release in man. Clin Endocrinol (Oxf). 1983;18:37-41.

72. Ostenson CG, Cattaneo AG, Doxey JC, Efendic S. Alpha-adrenoreceptors and insulin release from pancreatic islets of normal and diabetic rats. Am J Physiol. 1989;257(3 Pt 1):E439-E443.

73. Chobanian AV, Bakris GL, Black HR, et al. National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure; National High Blood Pressure Education Program Coordinating Committee. The sev-enth report of the Joint National Committee on prevention, Detection, evaluation, and treatment of high blood pres-sure: the JNC 7 report. JAMA. 2003;289:2560-2572.

74. Jiménez C, Cote G, Arnold A, Gagel RF. Review: should patients with apparently sporadic pheochromocytomas or paragangliomas be screened for hereditary syndromes? J Clin Endocrinol Metab. 2006;91:2851-2858.

75. Lefebvre M, Foulkes WD. Pheochromocytoma and para-ganglioma syndromes, genetics and management update. Curr Oncol. 2014;21:E8-E17.

Page 15: Sandi-Jo Galati, MD*; Meena Said, MD

382

76. Neumann HP, Bausch B, McWhinney SR, et al. Germ-line mutations in nonsyndromic pheochromocytoma. N Engl J Med. 2002;346:1459-1466.

77. Fishbein L, Merrill S, Fraker DL, Cohen DL, Nathanson KL. Inherited mutations in pheochromocytoma and para-ganglioma: why all patients should be offered testing. Ann Surg Oncol. 2013;20:1444-1450.

78. Giminez-Roqueplo AP, Dahia PL, Robledo M. An update on the genetics of paraganglioma, pheochromocy-toma, and associated hereditary syndromes. Horm Metab Res. 2012;44:328-333.

79. Rattenberry E, Vialard L, Yeung A, et al. A compre-hensive next generation sequencing based genetic test-ing strategy to improve diagnosis of inherited pheochro-mocytoma and paraganglioma. J Clin Endocrinol Metab. 2013;98:E1248-E1256.

80. Erlic Z, Rybicki L, Peczkowska M, et al. Clinical predic-tors and algorithm for genetic diagnosis of pheochromocy-toma patients. Clin Cancer Res. 2009;15:6378-6385.

81. Neary NM, King KS, Pacak K. Drugs and pheochromo-cytoma—don’t be fooled by every elevated metanephrines. N Engl J Med. 2011;364:2268-2270.

82. Lenders JW M, Willemsen JJ, Eisenhofer G, et al. Is supine rest necessary before blood sampling for plasma metanephrines? Clin Chem. 2007;53:352-354.

83. Grassi G, Seravalle G, Calhoun DA, Bolla G, Mancia G. Cigarette smoking and the adrenergic nervous system. Clin Exp Hypertens A. 1992;14:251-260.

84. Eisenhofer G, Huysmans F, Pacek K, Walther MM, Sweep FC, Lenders JW. Plasma metanephrines in renal failure. Kidney Int. 2005;67:668-677.