vasopressin: a review of therapeutic applications

18
REVIEW ARTICLE Vasopressin: A Review of Therapeutic Applications Natalie F. Holt, MD, MPH,*† and Kenneth L. Haspel, MD*† V ASOPRESSIN (VP) WAS DISCOVERED in 1895 from the extract of the posterior pituitary and named for the early observation of its vasoconstrictive properties. 1 The peptide is present in various species, both invertebrate and vertebrate, with only minor variations in amino acid se- quence. Human VP contains the amino acid arginine, hence the name arginine vasopressin. It is alternatively referred to as antidiuretic hormone (ADH), reflecting its main physio- logic function in promoting water retention. (For the remain- der of the text, the term vasopressin and the abbreviation VP will be used in lieu of arginine vasopressin or antidiuretic hormone.) Since its isolation 2 and synthetic preparation, 3 VP has been the subject of much research. Its clinical applications range from the management of enuresis to the treatment of gastrointestinal hemorrhage, septic shock, cardiac arrest, and heart failure. The purpose of this review is to provide an overview of the physiology and pharmacology of this hor- mone and its various functions. As with much in medicine, the discovery of disease states involving VP and/or its receptors has offered the most insight into VP’s in vivo role and functional repertoire. As such, an appraisal of these conditions will be used to review current knowledge and clinical applications of VP and its synthetic agonists and antagonists. Unanswered questions and future research op- portunities will then be summarized. VASOPRESSIN PHYSIOLOGY AND PHARMACOLOGY Vasopressin Synthesis and Regulation VP contains 9 amino acids, including 2 cysteines bridged by a disulphide bond. The gene for VP is on chromosome 2 and neighbors that of oxytocin. The 2 hormones are struc- tural cousins, differing only by 2 amino acids. 4,5 VP is synthesized in the magnocellular and parvocellular neurons of the hypothalamic paraventricular and supraoptic nuclei (Fig 1). It is transcribed as a larger prohormone with the carrier protein neurophysin, and it is cleaved into its final form while in transit via the supraoptic-hypophyseal tract to secretory granules in the posterior pituitary gland. 4 Upon stimulation, VP is released from its storage granules and rapidly enters the bloodstream via the well-vascularized posterior pituitary capillary bed, which is devoid of a blood- brain barrier. The most important stimulus for VP release is a change in plasma osmolality that is sensed via peripheral receptors near the portal vein and central receptors near the third ventricle. Informa- tion from peripheral receptors ascends via the vagus nerve through the medulla to the hypothalamic nuclei. The system is precisely regulated to maintain plasma osmolality between 285 and 295 mOsm/kg H 2 O. There is a linear relationship between plasma VP concentrations and plasma osmolality, and a change on the order of only 1 pg/mL in VP concentration produces a measureable adjustment in urine specific gravity. 6 VP is also responsive to changes in blood pressure, as sensed by baroreceptors in the left heart, aortic arch, and carotid sinus. Under ordinary physiologic conditions, VP has a limited role in blood pressure regulation. Evidence for this is in the lack of hypertensive effect seen among patients with the syndrome of inappropriate antidiuretic hormone secretion (SIADH), in which concentrations of VP are well above normal levels. 7 Whereas an alteration in plasma osmolality of only 1% effects a measurable change in plasma VP concentrations, blood pres- sure must drop by more than 10% to stimulate VP release. 6 However, with significant drops in blood pressure, plasma VP concentrations increase exponentially, in contrast to the linear VP response seen with changes in osmolality. 4 Other conditions have been noted to alter VP release either in vivo or in vitro; these include pain, hypoxemia, and hypogly- cemia. 8 Acetylcholine, histamine, catecholamines, prostaglan- dins, and nitric oxide (NO) appear to stimulate VP release, whereas opioids seem to inhibit VP release. 9 Furthermore, a role for VP has been postulated in human functions other than osmo- and baroregulation, including social behavior 10 and tem- perature regulation. 11 Normal plasma VP concentrations vary among individuals but are usually on the order of 1 to 5 pg/mL. Diurnal variation is observed, with a nighttime increase to approximately 2 times that of daytime levels. 12 The half-life of VP is between 5 and 20 minutes. 13 It is metabolized in the liver and kidneys. VP release triggers a number of negative feedback loops. The reduction in plasma osmolality inhibits VP synthesis. Adrenocorticotropin hormone (ACTH), whose release is stimulated by VP, increases glucocorticoid concentration, which inhibits VP secretion. In addition, VP induces a structural change in its own receptors, which inhibits their downstream interaction with G-proteins From the *Veterans Affairs Connecticut Healthcare System, West Haven Campus, West Haven, CT; and †Department of Anesthesiology, Yale University School of Medicine, New Haven, CT. Address reprint requests to Natalie F. Holt, MD, MPH, Veterans Affairs Connecticut Healthcare System, West Haven Campus, Depart- ment of Anesthesiology, 950 Campbell Avenue, West Haven, CT 06516. E-mail: [email protected] Published by Elsevier Inc. 1053-0770/10/2402-0022$36.00/0 doi:10.1053/j.jvca.2009.09.006 Key words: vasopressin, vaptans, arginine vasopressin, antidiuretic hormone, vasopressors 330 Journal of Cardiothoracic and Vascular Anesthesia, Vol 24, No 2 (April), 2010: pp 330-347

Upload: others

Post on 19-Feb-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Vepvqtaldwh

brgaomtraccap

V

batso(

HY

AmE

h

3

REVIEW ARTICLE

Vasopressin: A Review of Therapeutic Applications

Natalie F. Holt, MD, MPH,*† and Kenneth L. Haspel, MD*†

cfssrpb

ppttrmcoa

bUbhiwWasHcV

vcdwrop

bitmtphga

ASOPRESSIN (VP) WAS DISCOVERED in 1895 fromthe extract of the posterior pituitary and named for the

arly observation of its vasoconstrictive properties.1 Theeptide is present in various species, both invertebrate andertebrate, with only minor variations in amino acid se-uence. Human VP contains the amino acid arginine, hencehe name arginine vasopressin. It is alternatively referred tos antidiuretic hormone (ADH), reflecting its main physio-ogic function in promoting water retention. (For the remain-er of the text, the term vasopressin and the abbreviation VPill be used in lieu of arginine vasopressin or antidiureticormone.)Since its isolation2 and synthetic preparation,3 VP has

een the subject of much research. Its clinical applicationsange from the management of enuresis to the treatment ofastrointestinal hemorrhage, septic shock, cardiac arrest,nd heart failure. The purpose of this review is to provide anverview of the physiology and pharmacology of this hor-one and its various functions. As with much in medicine,

he discovery of disease states involving VP and/or itseceptors has offered the most insight into VP’s in vivo rolend functional repertoire. As such, an appraisal of theseonditions will be used to review current knowledge andlinical applications of VP and its synthetic agonists andntagonists. Unanswered questions and future research op-ortunities will then be summarized.

VASOPRESSIN PHYSIOLOGY AND PHARMACOLOGY

asopressin Synthesis and Regulation

VP contains 9 amino acids, including 2 cysteines bridgedy a disulphide bond. The gene for VP is on chromosome 2nd neighbors that of oxytocin. The 2 hormones are struc-ural cousins, differing only by 2 amino acids.4,5 VP isynthesized in the magnocellular and parvocellular neuronsf the hypothalamic paraventricular and supraoptic nucleiFig 1). It is transcribed as a larger prohormone with the

From the *Veterans Affairs Connecticut Healthcare System, Westaven Campus, West Haven, CT; and †Department of Anesthesiology,ale University School of Medicine, New Haven, CT.Address reprint requests to Natalie F. Holt, MD, MPH, Veterans

ffairs Connecticut Healthcare System, West Haven Campus, Depart-ent of Anesthesiology, 950 Campbell Avenue, West Haven, CT 06516.-mail: [email protected] by Elsevier Inc.1053-0770/10/2402-0022$36.00/0doi:10.1053/j.jvca.2009.09.006Key words: vasopressin, vaptans, arginine vasopressin, antidiuretic

wormone, vasopressors

30 Journal of Cardiothor

arrier protein neurophysin, and it is cleaved into its finalorm while in transit via the supraoptic-hypophyseal tract toecretory granules in the posterior pituitary gland.4 Upontimulation, VP is released from its storage granules andapidly enters the bloodstream via the well-vascularizedosterior pituitary capillary bed, which is devoid of a blood-rain barrier.The most important stimulus for VP release is a change in

lasma osmolality that is sensed via peripheral receptors near theortal vein and central receptors near the third ventricle. Informa-ion from peripheral receptors ascends via the vagus nerve throughhe medulla to the hypothalamic nuclei. The system is preciselyegulated to maintain plasma osmolality between 285 and 295Osm/kg H2O. There is a linear relationship between plasma VP

oncentrations and plasma osmolality, and a change on the orderf only 1 pg/mL in VP concentration produces a measureabledjustment in urine specific gravity.6

VP is also responsive to changes in blood pressure, as sensedy baroreceptors in the left heart, aortic arch, and carotid sinus.nder ordinary physiologic conditions, VP has a limited role inlood pressure regulation. Evidence for this is in the lack ofypertensive effect seen among patients with the syndrome ofnappropriate antidiuretic hormone secretion (SIADH), inhich concentrations of VP are well above normal levels.7

hereas an alteration in plasma osmolality of only 1% effectsmeasurable change in plasma VP concentrations, blood pres-

ure must drop by more than 10% to stimulate VP release.6

owever, with significant drops in blood pressure, plasma VPoncentrations increase exponentially, in contrast to the linearP response seen with changes in osmolality.4

Other conditions have been noted to alter VP release either inivo or in vitro; these include pain, hypoxemia, and hypogly-emia.8 Acetylcholine, histamine, catecholamines, prostaglan-ins, and nitric oxide (NO) appear to stimulate VP release,hereas opioids seem to inhibit VP release.9 Furthermore, a

ole for VP has been postulated in human functions other thansmo- and baroregulation, including social behavior10 and tem-erature regulation.11

Normal plasma VP concentrations vary among individualsut are usually on the order of 1 to 5 pg/mL. Diurnal variations observed, with a nighttime increase to approximately 2 timeshat of daytime levels.12 The half-life of VP is between 5 and 20inutes.13 It is metabolized in the liver and kidneys. VP release

riggers a number of negative feedback loops. The reduction inlasma osmolality inhibits VP synthesis. Adrenocorticotropinormone (ACTH), whose release is stimulated by VP, increaseslucocorticoid concentration, which inhibits VP secretion. Inddition, VP induces a structural change in its own receptors,

hich inhibits their downstream interaction with G-proteins

acic and Vascular Anesthesia, Vol 24, No 2 (April), 2010: pp 330-347

aAimpdrfc

VS

R

d(v

t

s

a r pitu

m ved.)

331VASOPRESSIN

nd results in cell sequestration of membrane-bound receptors.4

pproximately 10% to 20% of VP stores are available formmediate release in response to stimulation.14 Secretion di-

inishes with persistent stimulation, as suggested by the bi-hasic course of plasma VP concentrations in pathologic con-itions such as septic shock.15 Orally administered VP isapidly hydrolyzed by trypsin, rendering it inactive.16,17 There-ore, exogenous VP must be administered parenterally and by

Sup

Poste(neur

I

Arterial supplyto hypothalamus

Forebrainpathways

Brainpathw

Neurohypophysealtract

Blood bornesignals reachingSON and PVN

Herring bodies

Anterior lobe

Fig 1. VP is synthesized in the magnocellular and parvocellular

ranscribed as a larger prohormone with the carrier protein neur

upraoptic-hypophyseal tract to secretory granules in the posterior p

nd rapidly enters the bloodstream via the well-vascularized posterio

edical illustration used with permission of Elsevier. All rights reser

ontinuous infusion because of its short half-life. t

asopressin Receptor Distribution andignaling Pathways

eceptor Subtypes and Distribution

VP receptors are widely distributed in the body, and 3istinct receptor subtypes have been identified (Table 1). V1also referred to as V1a) receptors are found primarily onascular smooth muscle where they contribute to vasoconstric-

Origin of vasopressin

Cell ofsupraopticnucleus

Axonal transportof secretoryproduct

Posterior lobe

Fenestratedcapillary

ventriculareus

ic nucleus

beophysis)

ite of vasopressinbsorption

Venous drainageof posterior lobe

r hypophyseal artery

Neurosecretory Ending (posterior pituitary)

noxAnoxA

Pituicyte processes

Capillary

Endothelium

Mast cell

FibroblastNeurosecretoryvesicles

Collagen spaceBasement membrane

ns of the hypothalamic paraventricular and supraoptic nuclei. It is

sin, and it is cleaved into its final form while in transit via the

ry gland. Upon stimulation, VP is released from its storage granules

itary capillary bed, which is devoid of a blood-brain barrier. (Netter

(Color version of figure is available online.)

Paranucl

raopt

rior loohyp

Sa

nferio

stemays

neuro

ophy

ituita

ion, especially in vascular beds of the mesentery, skin, and

sptarmtemvpfiiatnhs

tciimWratVt

aTlopw

cmsbcmgntro

Vrgcrv

lgftnac

D

sigimpa

eptid

332 HOLT AND HASPEL

keletal tissues. They also are present on platelets where theyromote platelet aggregation. In the liver, V1-receptor activa-ion stimulates glycogenolysis and glucose secretion. In thedrenal gland, it promotes aldosterone and cortisol release. V1eceptors also have been found in the myometrium where theyediate uterine contractions. V1 receptors are selectively dis-

ributed in the kidney as well, such that activation constrictsfferent but not afferent arterioles.5 This effect appears to beediated by local release of NO18 and accounts for the obser-

ation that, although VP given to patients in shock statesromotes vasoconstriction, it also tends to increase glomerularltration rate and urinary output. Finally, there are V1 receptors

n the central nervous system, including the hypothalamus,mygdala, and cerebellum. V1 receptors in the brainstem seemo enhance baroreceptor-mediated inhibition of sympatheticervous system (SNS) activity;19,20 they also are postulated toave a role in other complex functions such as social memory,tress adaptation, and mood.21

Although V1 receptors are present in the kidney, V2 recep-ors predominate and are concentrated on cells of the distalonvoluted tubule and collecting duct. V2-receptor activationn the kidney stimulates gene transcription and cell membranentegration of aquaporin channels, which increase water per-eability at the luminal side of renal collecting duct cells.ater is then translocated to the basolateral cell surface and

eturned to the intravascular compartment. V2 receptors arelso expressed on the vascular endothelium, as evidenced byhe increased levels of von Willebrand factor (vWF), factorIII, and plasminogen activator that accompany the adminis-

ration of V2-receptor agonists.5,22

V3 (also called V1b) receptors have been identified in thenterior pituitary and elsewhere in the central nervous system.hrough these receptors, VP is believed to act as a neuromodu-

ator. It appears to stimulate ACTH secretion via the activationf the corticotropin-releasing hormone (CRH). This is sup-orted by the observation that exogenously administered CRH

Table 1. Distribution an

VP Receptor Subtype Location

V1 Vascular smooth muscle Vasoconstriction (Platelets Platelet aggregatiAdrenal gland Aldosterone and cLiver Glycogenolysis anGastrointestinal tract PeristalsisMyometrium Uterine contractioBrain Regulation of bloo

Temperature reguRole in emotional

Kidney Efferent arteriolarV2 Kidney Free water resorp

channels on renVascular smooth muscle VasodilationVascular endothelium Release of von W

V3 Anterior pituitary NeuromodulationBrain Stress adaptationPancreas Insulin synthesisHeart Atrial natriuretic p

ith VP result in substantially higher plasma cortisol levels d

ompared with the administration of CRH alone.13,23 Further-ore, during cardiopulmonary resuscitation (CPR) and early

epsis, short-term rises in serum ACTH and cortisol levels haveeen observed in patients treated with VP compared withatecholamines.24,25 V3-receptor activation also seems to pro-ote the secretion of other hormones, including prolactin,

rowth hormone, insulin, angiotensin, endothelin, and atrialatriuretic peptide (ANP), although the in vivo relevance ofhese properties remains to be established.26–29 Finally, V3eceptors have possible roles in social behavior and regulationf mood.21,30,31

Consistent with the observed molecular similarity betweenP and oxytocin, VP has been found to interact with oxytocin

eceptors, although with an affinity approximately one-tenth asreat as that of oxytocin. Recently, the interaction of VP withardiac oxytocin receptors has been found to promote ANPelease and subsequent natriuresis.32 The role of this system inivo has yet to be fully elucidated.VP also has activity at purinergic receptors, whose intrinsic

igand is the phosphate moiety of adenosine triphosphate.5 Inuinea pigs, VP infused into the coronary circulation has beenound to cause vasoconstriction via interaction at purinorecep-ors in the cardiac endothelium.33 However, these results haveot been reproduced consistently in vitro34 or animal models,35

nd the clinical significance of VP’s affinity for the purinore-eptor in humans remains to be shown.

ownstream Signaling

VP activity is mediated intracellularly through second mes-engers (Fig 2). V1 and V3 receptors act via a phosphatidyl-nositol pathway, producing inositol triphosphate and diacyl-lycerol, which, in turn, activate protein kinase C and increasentracellular calcium entry. In contrast, V2 receptor effects areediated by adenyl cyclase and cyclic adenosine monophos-

hate.12,36 As previously mentioned, other mechanisms for VPction, including locally induced NO release, also have been

ction of VP Receptors

Function

ntery, skin, skeletal tissue)

l releasecose release

ssure and heart rate

ing, social memory, circadian rhythm, stress adaptationtrictionia gene transcription and cell membrane integration of aquaporinllecting duct cells

nd factor, factor VIIIrticotrophin, growth hormone, prolactin secretion

leasee synthesis and release

d Fun

meseonortisod glu

nd prelationlearnconstion val co

illebra¡ co

and re

escribed.18

S

rorapt

T

wicrpflmtrihh

mcvwFspdtt

D

aV2cttDTolps

V

ht

s

p

t

V

m

m

r

F

c

o

m

u

b

a

a

a

h

d

c

l

333VASOPRESSIN

ynthetic Vasopressin Analogs

The development of VP analogs with longer half-lives andeceptor-specificity greatly enhanced the practical applicationf VP therapy. In 1968, octapressin was shown to improveenal blood flow in patients with cirrhosis.37 In 1985, Lenz etl38 found similar effects with another synthetic analog, orni-ressin. At present, the only clinically used VP analogs areerlipressin (TP) and desmopressin (DDAVP) (Fig 3).

P

TP (triglycyl lysine vasopressin) is a 12 amino acid peptidehose active metabolite is lysine vasopressin. Relative to VP,

t exhibits a greater degree of V1-receptor specificity (2.2:1ompared with 1:1 for AVP).39 Because lysine vasopressin iseleased into the systemic circulation gradually, when com-ared with VP, TP has a longer duration of action that allowsor bolus injection. However, because bolus dosing has beeninked to a higher frequency of systemic side effects, includingyocardial ischemia and skin necrosis, continuous infusion

herapy is favored.40,41 Like VP, TP is only available for pa-ental use. Of note, TP, but not VP, has been shown to dilatentrahepatic vessels, thereby enhancing blood flow through theepatic artery in patients with cirrhotic liver disease and portalypertension.42–44 TP has been available in Europe and Asia for

Fig 2. VP activity is mediated intracellularly through second mes-

engers. V1 and V3 receptors act via a phosphatidylinositol pathway,

roducing inositol triphosphate (IP3) and diacylglycerol (DAG), which in

urn activate protein kinase C and increase intracellular calcium entry.

2-receptor effects are mediated by adenyl cyclase and cyclic adenosine

onophosphate (cAMP). Receptor activation leads to synthesis and cell

embrane integration of aquaporin channels onto the luminal side of

enal collecting duct cells, which increases their permeability to water.

ree water is in turn translocated to the basolateral surface of renal

ollecting duct cells and returned to the central circulation. ATP, aden-

sine triphosphate; AVP, arginine vasopressin; cAMP, cyclic adenosine

onophosphate; DAG, diacylglycerol; Gq/Gs �, �, �, G protein sub-

mnits; IP3, inositol (1,4,5) trisphosphate; PIP2, phosphatidylinositol (4,5)-

isphosphate. (Reprinted with permission.13)

ore than 2 decades where it is considered the treatment ofhoice for the emergency management of bleeding esophagealarices. In addition, it is used “off-label” in treating patientsith refractory arterial hypotension. In 2004, the United Statesood and Drug Administration (FDA) granted TP orphan drugtatus as the only therapy being studied for the treatment ofatients with acute and rapidly progressive hepatorenal syn-rome (HRS); the following year, it was placed on the “fastrack” for FDA New Drug Approval where it remains at theime of this publication.45

DAVP

DDAVP (1-deamino-8-D-arginine vasopressin) was the firstnd remains the only clinically available VP agonist with2-receptor specificity (V2:V1 receptor affinity approximately,000-3,000 times that of VP).46 Intravenous, intranasal, sub-utaneous, and oral preparations of DDAVP are now standardreatment for patients with central diabetes insipidus (DI). Inhe 1970s, 2 independent research groups discovered thatDAVP increased the release of vWF and factor VIII.47– 49

his led to the use of prophylactic DDAVP to reduce peri-perative bleeding in patients with some types of von Wil-ebrand disease as well as hemophilia and uremia-inducedlatelet dysfunction (see section on Disorders of Hemosta-is).50 –52

asopressin Antagonists

Hyponatremia is one of the most common diagnoses inospitalized patients, occurring in the context of many condi-ions, including the syndrome of inappropriate antidiuretic hor-

Fig 3. VP contains 9 amino acids, including 2 cysteines bridged by

disulphide bond. The synthetic VP analogs desmopressin (DDAVP)

nd terlipressin (TP) are structurally similar to VP but differ slightly in

mino acid sequences. Relative to VP, the first amino acid of DDAVP

as been deaminated, and the arginine at the eighth position is in the

extro rather than the levo form. TP is a 12 amino acid prodrug

apable of slowly releasing lysine vasopressin, which accounts for its

onger half-life and V1-receptor specificity relative to vasopressin.

one (SIADH) secretion, congestive heart failure, and cirrho-

stfniCvSvaestepcd

D

isci(aDa

tn

dmgmCaorwDlpattfrtu

H

S

onF

to

334 HOLT AND HASPEL

is. Although fluid restriction and diuretics are often effective atreating hyponatremia, prolonged treatment may impair renalunction. In the 1990s, several nonpeptide vasopressin antago-ists, also called vaptans, were developed to oppose VP-nduced free water conservation present in these conditions.53

onivaptan is a parenterally formulated vaptan and the onlyaptan with substantial activity at both V1 and V2 receptors.everal other vaptans including tolvaptan, lixivaptan, moza-aptan, and satavaptan are specific V2-receptor antagonists andre available in oral form.54–57 All have shown therapeuticfficacy in terms of increasing urine output and raising serumodium concentrations. They are generally well tolerated, withhirst and dry mouth being among the most common sideffects.58 Vaptans are hepatically metabolized; conivaptan is aotent inhibitor of the CYP34A enzyme, and, as such, its usean alter the serum concentrations of many concomitantlyelivered drugs.58,59

VASOPRESSIN IN PATHOPHYSIOLOGIC CONDITIONS:DISORDERS OF PLASMA OSMOLALITY

I

DI is a condition characterized by the excess production ofnappropriately dilute urine. Loss of free water in excess ofolute leads to plasma hyperosmolality. The following 2 mainauses of DI are recognized: (1) a lack of VP secretion (pitu-tary or central DI) and 2) an impaired renal response to VPnephrogenic DI). Central DI is acquired and may occur frombrain tumor, infection, injury, or status postpituitary surgery.DAVP is now the mainstay of treatment for central DI. In

Table 2. Common Indications and Dosi

Indication

VP and Synthetic VP AnalogsCardiac arrestRefractory hypotension

Bleeding esophageal varices, hepatorenal syndrome

von Willebrand disease, hemophilia A, uremic platelet dysfun

Central diabetes insipidus

Abdominal distentionNocturnal enuresis

Vasopressin antagonistsHyponatremia, congestive heart failure, cirrhosis

Abbreviations: IV, intravenously; IM, intramuscularly; SC, subcutan*Current American Heart Association and European Resuscitation C

o epinephrine therapy in the management of pulseless cardiac arrest.f VP in refractory cases.137,138

ddition, DDAVP is used in the temporary treatment of noc- c

urnal enuresis, which is caused by a maturational delay in theormal nocturnal increase in VP secretion.Nephrogenic DI may be acquired or congenital. Genetic

efects in the V2 receptor, which is encoded on the X chro-osome, are responsible for most cases of congenital nephro-

enic DI. Defects in the aquaporin channel, encoded on chro-osome 12, also can cause congenital nephrogenic DI.21,60–62

ertain drugs, most notably lithium, interfere with the kidney’sbility to respond to VP and are responsible for acquired casesf nephrogenic DI. Relative to congenital cases, acquired neph-ogenic DI is usually of a milder form.21,63 Because patientsith nephrogenic DI do not respond appropriately to VP,DAVP is ineffective. To date, treatment has consisted of a

ow-sodium, low-protein diet; aggressive hydration; and theossible addition of nonsteroidal anti-inflammatory drugsnd/or diuretics, which, paradoxically, reduce urinary output inhese patients. Recent research indicates that V2-receptor an-agonists may help stabilize mutant VP receptors responsibleor some cases of nephrogenic DI and thereby improve theiresponsiveness to endogenous VP.64 However, investigations inhis area are still preliminary, and clinical application remainsnclear.

yponatremia

IADH

SIADH describes a condition in which there is dysregulationf VP secretion. The hallmark of SIADH is euvolemic hypo-atremia and urine osmolality in excess of plasma osmolality.irst described in the late 1950s, SIADH is one of the most

r VP, VP Analogs, and VP Antagonists

Drug Dose

VP 40 U IV � 1-2 dose(s)*VP 0.01-0.04 U/min IVTP 1-2 mg IV every 4-6 hours

0.1-0.4 mg/h IVVP 0.2-1.0 U/min IVTP 1-2 mg IV every 4-6 hoursDDAVP 0.3 �g/kg IV

300 �g intranasallyVP 5-10 U IM or SCDDAVP 5-40 �g intranasally

0.3-0.6 mg orally daily1-4 �g IV daily

VP 5-10 U IMDDAVP 5-20 �g intranasally

0.2-0.6 mg orally at bedtime

Conivaptan 20-80 mg IV dailyTolvaptan 15-90 mg orally dailyLixivaptan 25-250 mg orally twice daily

ly.il guidelines advise a single dose of intravenous VP as an alternativever, there is evidence to support the administration of a second dose

ng fo

ction

eousouncHowe

ommon diagnoses in hospitalized patients.65,66 It is observed in

ciscticc

flccmctpsm

toflapcspab

C

otoalpastah(

tppmpndpstiaro

cpET

pgnhaitcuaaecaipwe

eaIfsahcTuhpApmflaa

vo

tpaptbhho

335VASOPRESSIN

onjunction with a number of pathologic states, including HIVnfection and tumors that produce ectopic VP (eg, pulmonarymall-cell carcinoma). SIADH is also a common postoperativeondition in which pain may contribute to excess VP secre-ion.67 In addition, medications are known to cause SIADH,ncluding phenothiazines, tricyclic antidepressants, nicotine,arbamazepine, and antineoplastic drugs such as vincristine andyclophosphamide.4,68

Most cases of SIADH are temporary and may be managed byuid restriction and/or diuretic therapy. In more severe orhronic conditions, drug therapy may be required. Demeclocy-line, a tetracycline agent, was traditionally a preferred phar-acotherapeutic option. Demeclocycline interferes with the

ellular response to VP in the kidney by impairing the forma-ion of the second messenger cyclic adenosine monophos-hate.57,69 Lithium also interferes with the renal response to VPecretion, but side effects precluded its routine use as a treat-ent for SIADH.70,71

The development of VP antagonists has introduced a newreatment option for patients with severe cases of SIADH andther chronic illnesses that cause symptomatic hyponatremia oruid retention. Intravenous conivaptan is the only vaptan FDA-pproved for the treatment of euvolemic or hypervolemic hy-onatremia in hospitalized patients (Table 2). Treatment withonivaptan consistently increases urinary output and raiseserum sodium concentrations.57,72,73 At doses used in clinicalractice, there is no effect on systemic blood pressure, althought high doses hypotension may occur because of V1-receptorlockade.

ongestive Heart Failure and Cirrhosis

Hyponatremia has been found to be an independent predictorf major complications, rehospitalization, and mortality in pa-ients with congestive heart failure.74–81 In a post hoc analysisf data from the Evaluation Study of Congestive Heart Failurend Pulmonary Artery Catheterization Effectiveness trial, base-ine and persistent hyponatremia were found to be independentredictors of hospitalization for heart failure and mortality over6-month follow-up period.82 Among these patients, suppres-

ion of VP release that would be expected because of hypona-remia is overridden by baroreceptor activation in the aorticrch and carotid sinus resulting from low cardiac output. En-anced activity of the renin-angiotensin-aldosterone systemRAAS) also may promote VP secretion.72,78,83,84

Gheorghiade et al85 studied the impact of 3 doses ofolvaptan in a double-blind placebo-controlled trial of 254atients with chronic heart failure. Over the 25-day studyeriod, reductions in body weight and edema, as well as nor-alization of serum sodium concentration in hyponatremic

atients, were observed in patients treated with tolvaptan butot placebo. Tolvaptan therapy was well tolerated and pro-uced no appreciable changes in heart rate, systemic bloodressure, or renal function. Interestingly, only 6.3% of thetudy population had abnormal VP levels at baseline, andolvaptan treatment did not produce a statistically significantncrease in VP levels; therefore, tolvaptan’s therapeutic mech-nism of action has yet to be fully established. However, theesults of this trial have since been substantiated by several

ther large-scale, randomized, placebo-controlled studies, in- f

luding the Acute and Chronic Therapeutic Impact of Vaso-ressin Antagonist in Congestive Heart Failure trial86 and thefficacy of Vasopressin Antagonist in Heart Failure Study witholvaptan trial.87,88

Other trials of V2 antagonists have been conducted amongatients with hyponatremia from cirrhosis, SIADH, and con-estive heart failure. All have shown efficacy in terms oformalization of serum sodium concentration.74,75,86,89–93 Manyave reported symptomatic benefits including reduced edemand increased ease of breathing.82,86–88,94,95 One study identifiedmprovements in self-assessed mental health over a 30-dayreatment period.91,96 Whether serum sodium normalizationonfers benefit in terms of morbidity and mortality remainsncertain. In animal models of congestive heart failure, V1/V2ntagonist therapy has been shown to improve fluid regulationnd reduce cardiac hypertrophy.96–102 Studies in humans havexhibited mixed results. A few short-term trials withonivaptan103 and tolvaptan104 have shown reductions in righttrial and pulmonary artery occlusion pressures and modestlymproved left ventricular function. However, in a 12-weeklacebo-controlled trial of oral conivaptan therapy in patientsith class III heart failure, no impact on functional capacity or

xercise tolerance was shown.105

The Multicenter Evaluation of Tolvaptan Effect On Remod-ling trial studied the impact of long-term tolvaptan therapy ingroup of 240 patients with New York Heart Association class

I or III heart failure and ejection fractions �30%. In a 1-yearollow-up, tolvaptan-treated patients exhibited statistically non-ignificant increases in left ventricular ejection fraction; inddition, there was a trend toward lower rates of mortality andeart failure–related hospitalizations.106 However, in the Effi-acy of Vasopressin Antagonist in Heart Failure Study witholvaptan trial, the largest study to date, investigators werenable to identify improvement in heart function, physicalealth, or overall mortality over a median 9.9-month follow-uperiod.88 Patient recruitment is now underway for the BAL-NCE study, a phase III randomized multicenter double-blind,lacebo-controlled, parallel study of oral lixivaptan for theanagement of hyponatremia in patients hospitalized for heart

ailure.72 BALANCE will study the safety and efficacy of oralixivaptan for increasing serum sodium concentration and willssess the impact of therapy on rehospitalization as well asll-cause and cardiovascular mortality.

DISORDERS OF HEMOSTASIS

on Willebrand Disease and Disordersf Coagulation

In the 1970s, 2 research groups independently discoveredhat DDAVP enhanced the release of factor VIII, vWF, andlasminogen activator, thereby promoting platelet aggregationnd coagulation.47–49 This effect is mediated via V2 receptorsresent on the vascular endothelium.107 Mannucci et al48 werehe first to show the effectiveness of DDAVP in reducingleeding during dental extraction in a group of 25 patients withemophilia A or von Willebrand disease.48 Since then, DDAVPas achieved widespread use in the perioperative managementf patients with von Willebrand disease type 1, which results

rom a partial quantitative reduction in vWF. DDAVP is tran-

stdcbaraspbaal

P

DtiIcsmmcttaau

V

isBolmepawU

H

sstpRbfta

itot

fpvecvptttce

S

lfptslpic

addrVpVabr

epcniSatpaemdFbet

336 HOLT AND HASPEL

iently effective in most patients with von Willebrand diseaseypes 2A, 2M, and 2N, which are caused by qualitative vWFefects, although its prophylactic use in these patients is moreontroversial.51,52,108 Paradoxically, in patients with von Wille-rand disease type 2B, DDAVP may cause thrombocytopeniand thereby increase bleeding risk.52 DDAVP is also used toeduce perioperative bleeding in patients with hemophilia And other conditions associated with coagulation dysfunction,uch as uremia109–111 and cirrhosis.112–115 In addition, it hasroven effective at improving platelet function and reducinglood loss in patients treated with antiplatelet agents such asspirin and ticlopidine.112,116–119 The mechanism of this effectppears to involve more than stimulation of procoagulant re-ease but remains to be fully explained.120

erioperative Bleeding

In the 1980s, speculation was raised that prophylacticDAVP administration might help reduce blood loss in pa-

ients without intrinsic bleeding disorders who were undergo-ng complex surgical procedures at risk for large blood loss.nitial favorable results were reported by several groups in theontext of spinal fusion, major joint arthroplasty, and cardiacurgeries.118,120,121 However, subsequent studies and severaleta-analyses failed to reproduce these benefits.92,122–129 In aeta-analysis of 12 randomized controlled trials of DDAVP in

ardiac surgery, Cattaneo et al50 in fact found more than doublehe incidence of myocardial infarction in DDAVP-treated pa-ients compared with placebo-treated controls (4.4% v 1.6%),lthough the result was not statistically significant. Therefore,lthough DDAVP may help promote surgical hemostasis, itsse seems best reserved for select cases.50,92,127,130–132

ariceal Bleeding

In patients with cirrhotic liver disease, fibrotic tissue impairsntrahepatic blood flow, leading to the formation of dilatedplanchnic collateral vessels and portal venous hypertension.ecause of their high pressure and large shunt volume, rupturef these collaterals is often a lethal complication of advancediver disease. Both VP and TP are first-line therapies in theanagement of variceal hemorrhage because both have pow-

rful vasoconstrictive effects in splanchnic vascular beds. Com-ared with VP, TP appears to have a wider safety profile and islso the only drug that has shown a survival benefit comparedith placebo.133 However, TP remains unavailable in thenited States.

DISORDERS OF HYPOPERFUSION

epatorenal Syndrome

Patients with portal hypertension and cirrhosis develop atate of relative arterial hypotension caused by distention ofplanchnic blood vessels and underfilling of the central arterialree. This results in activation of the RAAS and SNS, whichromotes water and sodium retention and vasoconstriction.AAS activation and relative central hypovolemia impairlood flow to vital organs. Progressive renal failure originatingrom liver cirrhosis is one of the most difficult complications toreat and as such is associated with a high level of morbidity

nd mortality. Two forms of HRS are recognized; type I HRS d

s rapidly progressive and usually results in death within dayso weeks unless liver transplantation is performed. The coursef type II HRS is more variable; however, median survival is onhe order of 6 months.134

In addition to splanchnic vasoconstriction, TP has beenound to reduce portal venous pressure and blood flow throughortosystemic shunts as well as to dilate intrahepatic bloodessels, leading to increased hepatic blood volume. To thextent that the latter effect restores blood volume to the centralirculation, there is attenuation of RAAS and SNS-mediatedasoconstriction, which increases renal blood flow and helpsreserve renal function. TP is now under active investigation inhe management of patients with type I HRS as a bridge therapyo liver transplant. In clinical trials, treatment has been showno improve both renal and cardiovascular function and possiblyonfer survival benefit.133,135–139 Current evidence supportsarly intervention to maximize treatment benefits.

eptic Shock

Septic shock represents a pathologic state in which vasodi-ation results in hypoperfusion and ultimately end-organ dys-unction. Absolute and relative hypovolemia are frequentlyresent as a result of fever-induced evaporative and gastroin-estinal losses and fluid extravasation from the intravascularpace caused by capillary leak. In addition, pathologic vasodi-ation persists despite significant reductions in systemic bloodressure. Multiple mechanisms appear to mediate this response,ncluding increased NO production and activation of potassiumhannels that relax vascular smooth muscle.140–142

In the early phase of sepsis, VP levels are acutely elevatednd then decline rapidly as the condition progresses.143–148 Theepletion of endogenous VP appears to contribute to the vaso-ilation associated with advanced sepsis,149 and an inverseelationship between patient survival and endogenous serumP levels in the late stage of shock has been observed.150 As aotent secretagogue, endotoxin may deplete glandular stores ofP.151–153 High levels of circulating catecholamines and NO

lso have been shown to suppress VP release.154 Impairedaroreceptor-mediated VP secretion seems also to play aole.140,146

Landry et al146 were the first to report that the infusion ofxogenous VP in a series of 5 patients with septic shockroduced a measurable improvement in vascular tone and in-reased urinary output (Table 2).146 This is in contrast to theegligible hypertensive effect of exogenous VP when admin-stered to healthy, euvolemic adults.146 The Vasopressin andeptic Shock Trial was a multicenter, randomized trial aimed atssessing the impact of adding VP to norepinephrine therapy inreating patients with septic shock.155 The study involved 779atients. Adverse events were similar in both groups. Theuthors found no difference between groups in the primaryndpoint measure of 28-day mortality or in the frequency ofajor organ dysfunction. However, patients treated with VP

id exhibit increases in creatinine clearance and urinary output.urthermore, in subgroup analyses, VP exhibited treatmentenefit among patients with less severe sepsis (based on nor-pinephrine infusion �15 �g/min at enrollment) as well as inhose at risk of renal dysfunction (based on Brussels organ

ysfunction criteria, serum creatinine �2.0 mg/dL).155

rrtspstoppttTatetweptp

VOstirstlfii

oflaamapw2uhd0t

ciespt

Vmt

C

ldmtRbiapamvevs

tnbrktodotm

tavSic1islachcb

abweTata

337VASOPRESSIN

Acting on the knowledge that VP enhances corticotropinesponsiveness and that patients in shock states often exhibitelative adrenal insufficiency, Russell et al156 recently evaluatedhe impact of concomitant VP and corticosteroid therapy ineptic patients. In a post hoc analysis of data from the Vaso-ressin and Septic Shock Trial, the authors found a statisticallyignificant reduction in mortality at 28 days among patientsreated with the combination of corticosteroids and VP aspposed to corticosteroids and norepinephrine (35.9% v 44.7%,� 0.03). Equally interesting was the finding that among

atients who did not receive corticosteroid therapy, thosereated with VP had a higher rate of mortality compared withhose who received norepinephrine (33.7% v 21.3%, p � 0.06).he authors found that patients treated with corticosteroids hadsubstantially greater increase in plasma VP levels relative to

hose who did not receive steroid therapy, which they hypoth-sized might account for the difference in therapeutic responseo VP among study groups. Although corticosteroid treatmentas not randomized in this study, the results offer compelling

vidence of an interaction between corticosteroids and VP inatients with shock and imply the need for further studies onhe use of combination therapies in the management of theseatients.In Europe, research has been performed using the synthetic

P analog TP in the treatment of patients in septic shock.’Brien et al157 first reported the successful use of TP in 8

eptic patients with refractory hypotension despite tradi-ional therapies. Subsequent reports, including a random-zed, prospective trial, confirmed these findings.158 –160 Aecent trial has been conducted comparing continuous infu-ion TP with norepinephine or VP as first-line therapy in thereatment of patients with resuscitated septic shock.161 Pre-iminary reports suggest no harm and possibly some benefitrom TP treatment. A prospective, randomized clinical trials currently underway to identify the optimal TP dose for thisndication (TESTT-I).161,162

Data are extremely limited on the impact of VP or TP on theutcome of pediatric patients with septic shock. Based on theew published case reports, VP deficiency appears to play aesser role in the pathophysiology of septic shock in the pedi-tric population compared with adults; however, VP and itsnalogs have in select cases resulted in some benefit. Untilore research is performed, it seems reasonable to cautiously

dvise the use of VP or TP as rescue therapy in pediatricatients with refractory shock. Reported doses have variedidely. For TP, boluses as low as 7 �g/kg every 12 hours andto 20 �g/kg every 4 hours have been used; however, contin-

ous infusions using doses as high as 10 to 20 �g/kg/h alsoave been reported.21,163,164 When VP has been used, bolusoses of 0.01 to 0.3 U/kg or continuous infusions of 0.0002 to.004 �g/kg/min appear to be most common with dose titrationo effect.163,165,166

In summary, the preferred treatment of vasodilatory shockaused by sepsis remains unknown. Whether a single approachs appropriate for all patients is also unclear. Studies to datemphasize the value of early pharmacologic intervention toupport arterial blood pressure and organ perfusion, and theotential benefit of combination therapy is under active inves-

igation. Although more research remains to be done, low-dose m

P (0.01-0.04 U/min) is now commonly used in the manage-ent of septic patients and is endorsed by international sepsis

reatment guidelines.167

ardiac Arrest

Although epinephrine has been the mainstay of pharmaco-ogic therapy for cardiac arrest, longstanding concerns over therug’s adverse profile (eg, increased myocardial oxygen de-and, arrhythmogenic potential, and postresuscitation hyper-

ension) have motivated research into alternative therapies.ecognizing VP as an important vasoactive hormone in theody’s intrinsic neuroendocrine stress response, in the 1990s,nterest in the potential role of VP in the setting of cardiacrrest heightened. Lindner et al168,169 were the first to report thatatients in cardiac arrest exhibited high levels of circulating VPnd, furthermore, that substantially higher levels of the hor-one were present in survivors compared with nonsurvi-

ors.168,169 Subsequent animal studies suggested that thexogenous administration of VP in cardiac arrest improvedital organ perfusion, neurologic outcomes, and short-termurvival.170 –176

Studies in human cardiac arrest patients have not consis-ently shown benefit from VP therapy; however, there also hasot been convincing evidence of demonstrable harm.177 On thisasis, in its updated advanced cardiac life support guidelineseleased in 2000, the American Heart Association (AHA) ac-nowledged the use of 40 U of intravenous VP as an alternativeo either the first or second dose of epinephrine in the contextf pulseless ventricular fibrillation or ventricular tachycar-ia. In 2005, AHA guidelines were revised to support the usef VP in all cases of pulseless cardiac arrest. Guidelines ofhe European Resuscitation Council (ERC) echo this recom-endation (Table 2).178

As with research in the treatment of septic shock, interest inhe potential benefit of combination therapy to treat cardiacrrest soon followed. Initial studies suggested a possible sur-ival benefit with the combined use of epinephrine and VP.179

ubsequently, a large-scale randomized, controlled trial involv-ng nearly 3,000 patients with out-of-hospital cardiac arrestompared injection of 1 mg of epinephrine and 40 U of VP tomg of epinephrine and saline placebo. The study failed to

dentify a benefit from the addition of VP in any of the mea-ured endpoints, which included return of spontaneous circu-ation, neurologic outcome at discharge, and out-of-hospitalnd 1-year survival.180 More recently, a randomized, placebo-ontrolled study involving 100 patients with refractory in-ospital cardiac arrest found that the addition of intravenousorticosteroids to VP and epinephrine conferred a survivalenefit.181

Information regarding the use of VP in pediatric cardiacrrest is extremely limited. Some early reports suggested aenefit.182 However, a recent analysis identified worse outcomeith respect to return of spontaneous circulation but no differ-

nce in the rate of hospital discharge with the use of VP.183

hese results may be biased by the tendency for VP to be useds a “last resort” among patients with prolonged arrest andhose with more significant underlying pathology. Therefore,lthough VP may be part of the armamentarium for the treat-

ent of pediatric cardiac arrest, there is too little research at

tdoods

c(pwrmciftarwhaar

H

taihbwvmtsaapsbbhdiriuot

A

m(ct

Casscipsgabihsptibd

cgfsigsN

PH

sethpfteh

ccrbleslaVrmtCu

338 HOLT AND HASPEL

his time to endorse its routine use, and, as such, the use of VPoes not appear in the AHA’s algorithm for the managementf pulseless arrest in pediatric patients.184 However, basedn published reports, when used for this indication, VPoses of 0.4 U/kg and TP doses of 15 to 20 �g/kg have beenuggested.165

Research on treatments for cardiac arrest is fraught withomplications; lack of patient homogeneity and study settingeg, in-hospital v out-of-hospital arrest) complicate the com-arison of study results. Furthermore, it is difficult to judgehat amount of investigational research and/or clinical expe-

ience should constitute grounds for a change in routine patientanagement. In fact, because studies on VP in the setting of

ardiac arrest are mixed, the AHA and ERC’s decisions tonclude VP on advanced cardiac life support algorithms have inact been met with circumspection. For one, critics emphasizehe high cost of VP relative to epinephrine (a single VP dose ispproximately 15 times more expensive than that of epineph-ine) as well as the additional cost and complications associatedith arming emergency medical response personnel and in-ospital “crash carts” with both drugs.177 Revisions to the AHAnd ERC’s guidelines are expected within the next 2 to 3 years,nd it remains to be seen whether VP will keep its place on theesuscitation algorithms endorsed by these organizations.

emorrhagic Shock

Given VP’s known effectiveness in controlling gastrointes-inal hemorrhage, several researchers have applied VP or itsnalogs to the management of patients presenting with trauma-nduced hemorrhagic shock. Voelckel et al185,186 reported en-anced renal perfusion and postresuscitation hemodynamic sta-ility in pigs treated with conventional CPR plus VP comparedith CPR plus epinephrine after experimentally induced hypo-olemia and cardiac arrest. Subsequently, in a similar experi-ental model, the same group reported favorable results with

he use of VP as first-line therapy for uncontrolled hemorrhagichock when compared with either fluid or epinephrine therapylone.186,187 Several case reports have acknowledged a favor-ble impact with VP as a temporizing measure to support bloodressure during triage of trauma victims.147,188–190 However,ome research suggests that although VP may help restorelood pressure and reduce resuscitative fluid requirements, thisenefit may come at the expense of adverse metabolic andemodynamic consequences, including lactic acidemia and aecline in cardiac index.191 Currently, a multicenter, random-zed controlled trial, the Vasopressin in Traumatic Hemor-hagic Shock (VITRIS) trial, is being organized in Europe tonvestigate the impact of VP in the prehospital management ofncontrolled hemorrhagic shock. It is expected that the resultsf this trial will help form more definitive recommendations onhe use of VP in the context of hemorrhagic shock.192–194

nesthesia-Induced Hypotension

Under ordinary circumstances, arterial blood pressure isaintained through the complementary action of the (1) SNS,

2) RAAS, and (3) vasopressinergic response. VP’s role inorrecting hypotension is in essence an emergency mechanism

hat manifests when other systems fail or are overwhelmed. s

onventionally used agents for both general and neuraxialnesthesia alter the SNS response to hypotension. Evidenceuggests that epidural anesthesia also impairs the renin re-ponse to hypotension via the blockade of renal SNS dis-harge.195–197 Much of the hypotension induced by anestheticss catechol responsive, as shown by the effective use of vaso-ressors such as phenylephrine and ephedrine to treat anesthe-ia-induced vasodilation. However, clinical experience sug-ests that at least some of the hypotension induced bynesthesia is refractory to catecholamines. In dogs, sympatheticlockade caused by epidural anesthesia has been shown tonduce a compensatory rise in endogenous VP secretion. Inumans, the exogenous administration of VP has been useduccessfully to treat catechol-resistant hypotension.198 VP ap-ears to be particularly useful in treating hypotension in anes-hetized patients who also are treated with angiotensin-convert-ng enzyme (ACE) inhibitors or angiotensin receptor blockersecause these patients suffer from concomitant SNS and RAASysfunction.198–206

VP also has been used successfully in the management ofardiovascular collapse because of anaphylaxis occurring undereneral anesthesia. Schummer et al207,208 reported the success-ul use of VP for this indication in 7 patients whose hypoten-ion was refractory to conventional catecholamine treatmentncluding epinephrine and norepinephrine. The authors sug-ested that VP in this context may in addition to its vasocon-tricting effect also exert anti-inflammatory action by bluntingO production.

ostcardiotomy/Cardiopulmonary Bypass–Inducedypotension

It has been estimated that approximately 10% of cardiacurgery patients experience vasodilatory hypotension that is notxplained by primary myocardial dysfunction or sepsis.209 Ex-racorporeal circulation or cardiopulmonary bypass inducesemostatic derangements that make patients especially prone toostoperative blood loss. Relative depletion of coagulationactors and fibrinogen as well as platelet dysfunction contributeo this condition. Additionally, cytokine release induced byndothelial injury induces vasodilation that exacerbates theemodynamic impact of postoperative blood loss.210–212

Patients undergoing cardiac surgery also experience signifi-ant fluctuations in serum VP concentrations. The initiation ofardiopulmonary bypass (CPB) is associated with a dramaticise in serum VP levels; this is followed by a gradual return toaseline in the postoperative period.213–216 In some patients, VPevels are inappropriately low relative to the degree of postop-rative hypotension and contribute to refractory vasodilatoryhock. Risk factors for the development of post-CPB vasodi-atory hypotension include low presurgical ejection fractionnd the use of ACE inhibitors.209 In these patients, exogenousP infusion has been found to effectively increase mean arte-

ial pressure (MAP) and reduce catecholamine require-ents.209,217,218 Furthermore, in a double-blind, randomized

rial, prophylactic VP infusion begun before the initiation ofPB was shown to improve hemodynamic stability in patientsndergoing coronary artery bypass graft surgery or valvular

urgery who also were treated with ACE inhibitors.206 De-

cs

4fctnr

tiasinavfte

aruvrsigct

ntiat

O

arbboc

csVitmsaora

clo

O

cVttVrft

rugddpea

samae

V

S

breVivbEeggldsflirnsv

339VASOPRESSIN

reases in time to extubation and length of intensive care unittay also were observed.

Dunser et al219 investigated the impact of VP infusion among1 patients with catechol-resistant postcardiotomy shock. Theyound that VP infusion was associated with significant in-reases in MAP, left ventricular stroke work index, and sys-emic vascular resistance along with a decrease in heart rate ando change in cardiac index or stroke volume index.219 Theseesults have been substantiated by other studies.218,220

The unique benefits and risks of VP therapy to treat vasodila-ory hypotension remain to be fully established. Some groups havedentified a reduction in markers of myocardial ischemia associ-ted with VP infusion.205,221 In a swine model, at low flow ratesuch as those used during CPB, Mayr et al222 found that VPmproved coronary artery blood flow and did not increase coro-ary vascular resistance.223 However, high-dose VP therapy suchs that used to control bleeding has been known to cause coronaryasoconstriction and produce a substantial negative inotropic ef-ect.224,225 The use of VP in patients with cardiogenic as opposedo vasodilatory hypotension is likely to pose the most risk ofxacerbating myocardial dysfunction.

There is also evidence to suggest that VP therapy may conferrenal-protective effect. VP infusion initiated in patients with

efractory post-CPB hypotension has been shown to increaserine output,218 and the use of VP to treat hypotension after leftentricular assist device insertion has resulted in improvedecovery of renal function.226 On the other hand, VP is a potentplanchnic vasoconstrictor and may promote gastrointestinalschemia in at-risk patients. VP also increases platelet aggre-ation, which could promote thrombogenesis and impair mi-rocirculation. However, no data suggest that these complica-ions are common.

Several vasopressors in addition to VP, including norepi-ephrine, phenylephrine, and methylene blue, have been showno increase MAP in postcardiotomy hypotension. To date, theres insufficient evidence to suggest the use of one exclusivegent.227 However, existing data clearly place VP among theherapeutic options.

ther Shock States

Evidence from case reports has offered the use of VP or itsnalogs as rescue therapy in other conditions associated withefractory hypotension including overdoses of calcium channellockers228 and tricyclic antidepressants229 as well as traumaticrain injury.230 By virtue of their sporadic occurrence, evidencef the efficacy of VP and VP analogs for these unusual cir-umstances will remain anecdotal.

The explanation for why VP successfully reverses cate-holamine-refractory vasodilation is not firmly established, buteveral hypotheses have been offered. Both catecholamines andP exert some of their vasoconstrictive effects by increasing

ntracellular calcium in vascular smooth muscle cells. In addi-ion, however, VP blocks cyclic guanosine monophosphate-ediated vasodilation produced by inflammatory mediators

uch as interleukin 1, ANP, and NO.209,231–233 VP also inhibitsdenosine triphosphate–mediated potassium channel activationn vascular smooth muscle cells, which improves vascularesponsiveness to catecholamine therapy.234–238 Vascular tone

lso may be restored by virtue of VP-mediated release of p

ortisol.239 The existence of these multimodal mechanismsikely explains the effectiveness of VP therapy in shock statesf various etiologies.

ther Applications

Several nonpeptide VP antagonists are in various stages oflinical trials for a wide assortment of clinical indications.1-receptor antagonists including relcovaptan are under inves-

igation for the management of Raynaud’s disease,240 prema-ure labor,241,242 dysmenorrhea,243,244 and Meniere’s disease.245

1-receptor antagonists also are offering new potential ineducing cortisol levels in patients with ACTH-independentorms of Cushing syndrome caused by aberrant adrenocor-ical receptors.246

The finding that cyclic adenosine monophosphate promotesenal epithelial cell growth has prompted investigations on thetility of V2-receptor antagonist therapy for slowing the pro-ression of renal dysfunction in patients with polycystic kidneyisease.247–255 Studies of V2-receptor antagonists for other in-ications including the reduction of intraocular pressure inatients with glaucoma256,257 and the minimization of cerebraldema and infarct size after ischemic brain injury256,258–261 arelso underway.

In the neuropsychiatric arena, V3 antagonist therapy hashown promise in reducing stress-induced cortisol secretionnd exerting anxiolytic and mood-elevating effects in rodentodels.262–277 SSR-149415, the only orally active V3-receptor

ntagonist, is currently in phase II clinical trials to evaluate itsfficacy in humans (Table 2).59,272

ADVERSE EFFECTS OF DRUG THERAPY

P and VP Analogs

planchnic Perfusion

Despite its beneficial role, significant adverse effects haveeen reported with both VP and its analogs. Although VPedistributes blood flow to vital organs, it does so in part at thexpense of splanchnic and peripheral tissue perfusion. As such,P has been observed to induce skin necrosis and severe limb

schemia, particularly when administered through peripheraleins.162,278–282 A decrease in bile flow and increases in biliru-in and transaminase levels also have been reported.158,220,223,283

xperimental models have produced conflicting results on theffect of VP or V1 agonists on gut microcirculation; someroups have measured an increase in the mucosal-arterial pCO2

radient, implying the potential for impaired mesenteric circu-ation.24,278,283–286 However, this result has not been consistentlyuplicated.285,287 The potentially deleterious effect of VP onplanchnic perfusion seems keenly influenced by the overalluid status of the patient when therapy is initiated; VP admin-

stered in the context of hypovolemia appears particularly det-imental, whereas VP in fluid-resuscitated states may result inet improvement in splanchnic hemodynamics.288 There areimilar conflicting reports on the effects of VP on coronaryessels; but acute myocardial ischemia has been observed

articularly with bolus dosing of TP.224,289

C

ripvbsvapetltrnf

H

ctamctcru

F

rVio

t

sAcvVrda

V

aaabtbtrd

hsvr

wiVvmdstcp

p2

c

pJ

e

v

od

a7

340 HOLT AND HASPEL

ardiac Index

Concern also has been raised that VP may worsen post-esuscitation acidemia and increase the risk of reperfusionnjury-induced multiorgan system dysfunction, despite im-roving arterial blood pressure and limiting resuscitative fluidolume requirements.191,192 Although VP therapy may restorelood pressure, depression of cardiac index has been ob-erved.24,146,158–160,203,223,286,290 However, this impact seems toary based on initial cardiac index. Specifically, Luckner etl223 showed that although VP decreased the cardiac index inatients with initially elevated cardiac indices, it had littleffect in patients whose cardiac index was normal at the start ofherapy and actually improved cardiac indices in patients withow starting values. This has led investigators to speculate thathe change in cardiac index observed with VP therapy mayepresent an appropriate adaptive response to blood pressureormalization rather than a pathologic effect on myocardialunction.40

emostasis

The activation of V1 receptors increases intracellular cal-ium and promotes platelet aggregation. V2-receptor stimula-ion promotes coagulation via release of vWF and plasminogenctivator. Theoretically, these hemostatic effects may compro-ise microcirculation, promote thromboembolism, and/or

ause bleeding complications related to thrombocytopenia. Al-hough some groups have observed a drop in platelet count withontinuous VP infusion and TP bolus therapy, thromboelastog-aphy, when performed, has failed to uncover functional coag-lation impairment.158,280,223,283,291,292

luid and Electrolyte Balance

Although VP is known to possess antidiuretic properties via V2eceptor activation in the kidneys, in the setting of septic shock,P and VP analog therapy consistently have been shown to

ncrease urinary output.140,155 Hyponatremia is a rare event andnly seems to be of concern when high-dose therapy is used.289

In summary, the pathophysiology of shock and the tradi-

ional catecholamine therapy used to treat it often entail organ- r

REN

1:173-178, 1986

h

t5

nm

o

p

c

r

p

pecific compromise; as such, patient prognosis is grim a priori.lthough conflicting reports have been published, there is no

onvincing evidence from human studies to suggest that ad-erse consequences occur in excess among patients treated withP or its analogs. Therefore, although definitive guidelines

emain forthcoming, VP and VP analog therapy have earnedeserved roles in the treatment armamentarium of catechol-mine-refractory shock.

P Antagonists

Relatively few adverse effects have been identified with VPntagonist treatment. In animal models, conivaptan has beenssociated with fetal malformations at subtherapeutic levelsnd has delayed labor onset in pregnant rats. It is therefore toe avoided in pregnancy. Conivaptan also substantially altershe concentrations of many concomitantly administered drugsecause of its powerful inhibition of the CYP3A4 enzyme. Forhis reason, it is only available in parenteral form, and theecommended duration of therapy is limited to 4 consecutiveays.56,59,73,97

Thirst, dry mouth, polyuria, and overrapid correction ofyponatremia are among the more common side effects ob-erved with the vaptans. They are usually only clinically rele-ant when high-dose therapy is used in conjunction with fluidestriction.53,57,59

CONCLUSION

Vasopressin is a homeostatic neuroendocrine hormonehose chief role is to maintain plasma osmolality. Although

mportant in health, in the study of disease states, the breadth ofP’s functional repertoire, now known to include regulation ofascular tone, alteration of blood coagulability, modulation ofood, and stress activation, has become apparent. Relative VP

epletion or abundance has been shown in multiple pathologictates, making VP agonist and antagonist therapies excitingargets of study in fields as disparate as psychiatry and criticalare medicine. The extent to which these therapies may im-rove patient outcomes and alter the natural history of disease

emain questions for ongoing research.

REFE

1. Oliver G, Schaefer E: On the physiological action of extract ofituitary body and certain other glandular organs. J Physiol 18:277-79, 1895

2. Dekanski J: The quantitative assay of vasopressin. Br J Pharma-ol Chemother 7:567-572, 1952

3. Du Vigneaud V, Gish D, Katsoyannis P: A synthetic preparationossessing biological properties associated with arginine-vasopressin.Am Chem Soc 76:4751-4752, 1954

4. Vincent J, Su F: Physiology and pathophysiology of the vasopressin-rgic system. Best Pract Res Clin Anaesthesiol 22:243-252, 2008

5. Maybauer M, Maybauer D, Enkhbaatar P, et al: Physiology of theasopressin receptors. Best Pract Res Clin Anaesthesiol 22:253-263, 2008

6. Bankir L: Antidiuretic action of vasopressin: Quantitative aspectsf interaction between V1a and AV3 receptor-mediated effects. Car-iovasc Res 51:372-390, 2001

7. Williams T, Da Costa D, Mathias C, et al: Pressor effect ofrginine vasopressin in progressive autonomic failure. Clin Sci (Lond)

CES

8. Baylis P: Osmoregulation and control of vasopressin secretion inealthy humans. Am J Physiol 253:R671-R678, 19879. Leng G, Brown C, Russell J: Physiological pathways regulating

he activity of magnocellular neurosecretory cells. Prog Neurobiol7:625-655, 199910. Bielsky I, Hu S, Ren X, et al: The V1a vasopressin receptor is

ecessary and sufficient for normal social recognition: A gene replace-ent study. Neuron 47:503-513, 200511. Weingartner H, Gold P, Ballenger J, et al: Effects of vasopressin

n human memory functions. Science 211:601-603, 198112. George C, Messerli F, Genest J, et al: Diurnal variation of

lasma vasopressin in man. J Clin Endocrinol Metab 41:332-338, 197513. Treschan TA, Peters J: The vasopressin system: Physiology and

linical strategies. Anesthesiology 105:599-612, 200614. Holmes C, Patel B, Russell J, et al: Physiology of vasopressin

elevant to management of septic shock. Chest 120:989-1002, 200115. Sklar A, Schrier R: Central nervous system mediators of vaso-

ressin release. Physiol Rev 63:1243-1280, 1983

d

pE

iv

sC

s2

s

m

vs

rI

mt3

aca

a

r1

th

tJ

ao

ah

ae

pP

cH

wi

v1

fG

tR

ta

o

tS

ph

dJ

sw2

1

oH

vh

aIB

da7

po4

c

aH

oV

t

sn

2

al

341VASOPRESSIN

16. Baumann G, Dingman J: Distribution, blood transport, andegradation of antidiuretic hormone. J Clin Invest 57:1109-1116, 197617. Beardwell C, Geelen G, Palmer H, et al: Radioimmunoassay of

lasma vasopressin in physiological and pathological states in man. Jndocrinol 67:189-202, 197518. Aki Y, Tamaki T, Kiyomoto H, et al: Nitric oxide may partic-

pate in V2 vasopressin-receptor-mediated renal vasodilation. J Cardio-asc Pharmacol 23:331-333, 199419. Hasser E, Cunnningham J, Sullivan M, et al: Area postrema and

ympathetic nervous system effects of vasopressin and angiotensin II.lin Exp Pharmacol Physiol 27:432-436, 200020. Grindstaff R, Cunningham J: Cardiovascular regulation of va-

opressin neurons in the supraoptic nucleus. Exp Neurol 171:219-226,00121. Frank E, Landgraf R: The vasopressin system—From antidiure-

is to psychopathology. Eur J Pharmacol 583:226-242, 200822. Mannucci PM: Desmopressin: an historical introduction. Hae-ophilia 14:1-4, 2008 (suppl 1)23. Thibonnier M, Preston J, Dulin N, et al: The human V3 pituitary

asopressin receptor: Ligand binding profile and density-dependentignaling pathways. Endocrinology 138:4109-4122, 1997

24. Lauzier F, Levy B, Lamarre P, et al: Vasopressin or norepineph-ine in early hyperdynamic septic shock: A randomized clinical trial.ntensive Care Med 32:1782-1789, 2006

25. Kornberger E, Prengel AW, Krismer A, et al: Vasopressin-ediated adrenocorticotropin release increases plasma cortisol concen-

rations during cardiopulmonary resuscitation. Crit Care Med 28:3517-521, 200026. Katoh K, Yoshida M, Kobayashi Y, et al: Responses induced by

rginine-vasopressin injection in the plasma concentrations of adreno-orticotropic hormone, cortisol, growth hormone and metabolitesround weaning time in goats. J Endocrinol 187:249-256, 2005

27. Cogan E, Debiève M, Philipart I, et al: High plasma levels oftrial natriuretic factor in SIADH. N Engl J Med 314:1258-1259, 1986

28. Meller W, Lewis D, Gehris T, et al: Human anterior pituitaryesponse to exogenous arginine vasopressin. Acta Endocrinol (Copenh)25:378-384, 199129. Balakrishnan S, Gopalakrishnan V, McNeill J: Endothelin con-

ributes to the hemodynamic effects of vasopressin in spontaneousypertension. Eur J Pharmacol 334:55-60, 199730. Dinan TG, Scott LV: Anatomy of melancholia: Focus on hypo-

halamic-pituitary-adrenal axis overactivity and the role of vasopressin.Anat 207:259-264, 200531. Scott L, Dinan T: Vasopressin and the regulation of hypothal-

mic-pituitary-adrenal axis function: Implications for the pathophysi-logy of depression. Life Sci 62:1985-1998, 199832. Gutkowska J, Jankowski M, Lambert C, et al: Oxytocin releases

trial natriuretic peptide by combining with oxytocin receptors in theeart. Proc Natl Acad Sci U S A 94:11704-11709, 199733. Zenteno-Savin T, Sada-Ovalle I, Ceballos G, et al: Effects of

rginine vasopressin in the heart are mediated by specific intravascularndothelial receptors. Eur J Pharmacol 410:15-23, 2000

34. Boarder M, Weisman G, Turner J, et al: G protein-coupled P2urinoceptors: From molecular biology to functional responses. Trendsharmacol Sci 16:133-139, 199535. Mei Q, Liang B: P2 purinergic receptor activation enhances

ardiac contractility in isolated rat and mouse hearts. Am J Physioleart Circ Physiol 281:H334-H341, 200136. Nadal M: Secretory rhythm of vasopressin in healthy subjects

ith inversed sleep-wake cycle: Evidence for the existence of anntrinsic regulation. Eur J Endocrinol 134:174-176, 1996

37. Cohn J, Tristani F, Khatri I: Systemic vasoconstrictor and renalasodilator effects of PLV-2 (octapressin) in man. Circulation 38:151-

57, 1968 p

38. Lenz K, Druml W, Kleinberger G, et al: Enhancement of renalunction with ornipressin in a patient with decompensated cirrhosis.ut 26:1385-1386, 198539. Nilsson G, Lindblom P, Ohlin M, et al: Pharmacokinetics of

erlipressin after single IV doses to healthy volunteers. Drugs Exp Clines 16:307-314, 199040. Ertmer C, Rehberg S, Westphal M: Vasopressin analogues in the

reatment of shock states: potential pitfalls. Best Pract Res Clin An-esthesiol 22:393-406, 2008

41. Singer M: Arginine vasopressin vs. terlipressin in the treatmentf shock states. Best Pract Res Clin Anaesthesiol 22:359-368, 200842. Kiszka-Kanowitz M, Henriksen J, Hansen E, et al: Effect of

erlipressin on blood volume distribution in patients with cirrhosis.cand J Gastroenterol 39:486-492, 200443. Blei A, Groszmann R, Gusberg R, et al: Comparison of vaso-

ressin and triglycyl-lysine vasopressin on splanchnic and systemicemodynamics in dogs. Dig Dis Sci 25:688-694, 198044. Hansen E, Strandberg C, Højgaard L, et al: Splanchnic haemo-

ynamics after intravenous terlipressin in anaesthetised healthy pigs.Hepatol 30:503-510, 199945. Fast track designation for terlipressin in type 1 hepatorenal

yndrome. Pharmaceutical News 2005 Apr 20. Available at: http://ww.news-medical.net/news/2005/04/20/9386.aspx. Accessed April0, 200946. Richardson D, Robinson A: Desmopressin. Ann Intern Med

03:228-239, 198547. Mannucci P, Aberg M, Nilsson I, et al: Mechanism of plasmin-

gen activator and factor VIII increase after vasoactive drugs. Br Jaematol 30:81-93, 197548. Mannucci P, Ruggeri Z, Pareti F, et al: 1-deamino-8-D-arginine

asopressin: A new pharmacological approach to the management ofaemophilia and von Willebrands’ diseases. Lancet 1:869-872, 197749. Cash J, Gader A, da Costa J: The release of plasminogen

ctivator and factor VIII to lysine vasopressin, arginine vasopressin,-desamino-8-D-argnine vasopressin, angiotensin and oxytocin in man.r J Haematol 27:373-374, 197450. Cattaneo M, Harris AS, Stromberg U, et al: The effect of

esmopressin on reducing blood loss in cardiac surgery—A meta-nalysis of double-blind, placebo-controlled trials. Thromb Haemost4:1064-1070, 199551. Sadler JE, Budde U, Eikenboom JC, et al: Update on the

athophysiology and classification of von Willebrand disease: A reportf the Subcommittee on von Willebrand Factor. J Thromb Haemost:2103-2114, 200652. Franchini M: The use of desmopressin as a hemostatic agent: A

oncise review. Am J Hematol 82:731-735, 200753. Farmakis D, Filippatos G, Kremastinos DT, et al: Vasopressin

nd vasopressin antagonists in heart failure and hyponatremia. Curreart Fail Rep 5:91-96, 200854. Serradeil-Le Gal C, Lacour C, Valette G, et al: Characterization

f SR 121463A, a highly potent and selective, orally active vasopressin2 receptor antagonist. J Clin Invest 98:2729-2738, 199655. Mitchell S, Hunter J: Vasopressin and its antagonists: What are

heir roles in acute medical care? Br J Anaesth 99:154-158, 200756. Decaux G: Long-term treatment of patients with inappropriate

ecretion of antidiuretic hormone by the vasopressin receptor antago-ist conivaptan, urea, or furosemide. Am J Med 110:582-584, 200157. Gross P: Treatment of hyponatremia. Intern Med 47:885-891,

00858. Finley JJ, Konstam MA, Udelson JE: Arginine vasopressin

ntagonists for the treatment of heart failure and hyponatremia. Circu-ation 118:410-421, 2008

59. Decaux G, Soupart A, Vassart G: Non-peptide arginine-vaso-

ressin antagonists: The vaptans. Lancet 371:1624-1632, 2008

n

ts

h

N3

tM7

c

ss

p(

N

os

s2

t5

t

EawM

oJ

f

an

A

cs

rE

ph

ac

p

h2

vr1

sn

rR2

te1

ihJ

tE

mmp1

ss4

oac

(n3

ev

aht

otO2

piT

rV

v

342 HOLT AND HASPEL

60. Bichet DG: Nephrogenic diabetes insipidus. Adv Chronic Kid-ey Dis 13:96-104, 200661. Spanakis E, Milord E, Gragnoli C: AVPR2 variants and muta-

ions in nephrogenic diabetes insipidus: Review and missense mutationignificance. J Cell Physiol 217:605-617, 2008

62. Birnbaumer M: The V2 vasopressin receptor mutations and fluidomeostasis. Cardiovasc Res 51:409-415, 200163. Peters H, Robben J, Deen P, et al: Water in health and disease:

ew aspects of disturbances in water metabolism. Neth J Med 65:325-32, 200764. Morello C, Cranmer L, Spector D: Suppression of murine cy-

omegalovirus (MCMV) replication with a DNA vaccine encodingCMV M84 (a homolog of human cytomegalovirus pp65). J Virol

4:3696-3708, 200065. Bartter FC, Schwartz WB: The syndrome of inappropriate se-

retion of antidiuretic hormone. Am J Med 42:790-806, 196766. Schwartz W, Bennett W, Curelop S, et al: A syndrome of renal

odium loss and hyponatremia probably resulting from inappropriateecretion of antidiuretic hormone. Am J Med 23:529-542, 1957

67. Kendler KS, Weitzman RE, Fisher DA: The effect of pain onlasma arginine vasopressin concentrations in man. Clin EndocrinolOxf) 8:89-94, 1978

68. Moritz M, Ayus J: The syndrome of inappropriate antidiuresis.Engl J Med 357:942, 200769. Forrest JJ, Cox M, Hong C, et al: Superiority of demeclocycline

ver lithium in the treatment of chronic syndrome of inappropriateecretion of antidiuretic hormone. N Engl J Med 298:173-177, 1978

70. White M, Fetner C: Treatment of the syndrome of inappropriateecretion of antidiuretic hormone with lithium carbonate. N Engl J Med92:390-392, 197571. Juurlink D, Mamdani M, Kopp A, et al: Drug-induced lithium

oxicity in the elderly: A population-based study. J Am Geriatr Soc2:794-798, 200472. Oghlakian G, Klapholz M: Vasopressin and vasopressin recep-

or antagonists in heart failure. Cardiol Rev 17:10-15, 200973. Ghali J, Koren M, Taylor J, et al: Conivaptan study group.

fficacy and safety of oral Conivaptan: A V1a/V2 vasopressin receptorntagonist, assessed in a randomized placebo-controlled trial in patientsith euvolemic or hypervolemic hyponatremia. J Clin Endocrinoletab 91:2145-2152, 200674. Schrier R, Gross P, Gheorghiade M, et al: Tolvaptan, a selective

ral vasopressin V2-receptor antagonist, for hyponatremia. N EnglMed 355:2099-2112, 200675. Lee C, Watkins M, Patterson J, et al: Vasopressin: A new target

or the treatment of heart failure. Am Heart J 146:9-18, 200376. Bakris G, Kusmirek S, Smith A, et al: Calcium antagonism

bolishes the antipressor action of vasopressin (V1) receptor antago-ism. Am J Hypertens 10:1153-1158, 199777. Francis G: Neuroendocrine activity in congestive heart failure.

m J Cardiol 66:33D-38D; discussion 38D-39D, 199078. Gavras H: Role of vasopressin in clinical hypertension and

ongestive cardiac failure: Interaction with the sympathetic nervousystem. Clin Chem 37:1828-1830, 1991

79. Rouleau J, Packer M, Moyé L, et al: Prognostic value of neu-ohumoral activation in patients with an acute myocardial infarction:ffect of captopril. J Am Coll Cardiol 24:583-591, 199480. Goldsmith S, Francis G, Levine T, et al: Impaired response of

lasma vasopressin to orthostatic stress in patients with congestiveeart failure. J Am Coll Cardiol 2:1080-1083, 198381. Szatalowicz V, Arnold P, Chaimovitz C, et al: Radioimmuno-

ssay of plasma arginine vasopressin in hyponatremic patients withongestive heart failure. N Engl J Med 305:263-266, 1981

82. Gheorghiade M, Rossi J, Cotts W, et al: Characterization and

rognostic value of persistent hyponatremia in patients with severe s

eart failure in the ESCAPE Trial. Arch Intern Med 167:1998-2005,00783. Khokhar A, Slater J, Ma J, et al: The cardiovascular effect of

asopressin in relation to its plasma concentration in man and itselevance to high blood pressure. Clin Endocrinol (Oxf) 13:259-266,98084. Gavras H, Hatzinikolaou P, North W, et al: Interaction of the

ympathetic nervous system with vasopressin and renin in the mainte-ance of blood pressure. Hypertension 4:400-405, 198285. Gheorghiade M, Niazi I, Ouyang J, et al: Vasopressin V2-

eceptor blockade with tolvaptan in patients with chronic heart failure:esults from a double-blind, randomized trial. Circulation 107:2690-696, 200386. Gheorghiade M, Gattis W, O’Connor C, et al: Effects of

olvaptan, a vasopressin antagonist, in patients hospitalized with wors-ning heart failure: A randomized controlled trial. JAMA 291:1963-971, 200487. Gheorghiade M, Konstam M, Burnett JJ, et al: Short-term clin-

cal effects of tolvaptan, an oral vasopressin antagonist, in patientsospitalized for heart failure: The EVEREST Clinical Status Trials.AMA 297:1332-1343, 2007

88. Konstam M, Gheorghiade M, Burnett JJ, et al: Effects of oralolvaptan in patients hospitalized for worsening heart failure: TheVEREST Outcome Trial. JAMA 297:1319-1331, 200789. Soupart A, Gross P, Legros J, et al: Successful long-term treat-ent of hyponatremia in syndrome of inappropriate antidiuretic hor-one secretion with satavaptan (SR121463B), an orally active non-

eptide vasopressin V2-receptor antagonist. Clin J Am Soc Nephrol:1154-1160, 200690. Ginès P, Wong F, Watson H, et al: Effects of satavaptan, a

elective vasopressin V(2) receptor antagonist, on ascites and serumodium in cirrhosis with hyponatremia: A randomized trial. Hepatology8:204-213, 200891. Abraham W, Shamshirsaz A, Mefann K, et al: Aquaretic effect

f Lixivaptan, an oral, non-peptide, selective V2-receptor vasopressinntagonist, in New York Heart Association functional class II and IIIhronic heart failure patients. J Am Coll Cardiol 47:1615-1621, 2006

92. Wong F, Blei A, Blendis L, et al: A vasopressin receptor antagonistVPA-985) improves serum sodium concentration in patients with hypo-atremia: A multicenter, randomized, placebo-controlled trial. Hepatology7:182-191, 2003

93. Zelster D, Rosansky S, VanRensburg H, et al: Assessment of thefficacy and safety of intravenous Conivaptan in euvolemic and hyper-olemic hyponatremia. Am J Nephrol 27:447-457, 200794. Gheorghiade M, Abraham W, Albert N, et al: Relationship between

dmission serum sodium concentration and clinical outcomes in patientsospitalized for heart failure: An analysis from the OPTIMIZE-HF regis-ry. Eur Heart J 28:980-988, 2007

95. Gheorghiade M, Orlandi C, Burnett J, et al: Rationale and designf the multicenter, randomized, double-blind, placebo-controlled studyo evaluate the Efficacy of Vasopressin antagonism in Heart Failure:utcome Study with Tolvaptan (EVEREST). J Card Fail 11:260-269,00596. Bishara B, Shiekh H, Karram T, et al: Effects of novel vaso-

ressin receptor antagonists on renal function and cardiac hypertrophyn rats with experimental congestive heart failure. J Pharmacol Expher 326:414-422, 200897. Tahara A, Saito M, Sugimoto T, et al: AVP-induced mitogenic

esponses of Chinese hamster ovary cells expressing human V1A or1B receptors. Pflugers Arch 437:219-226, 199998. Tahara A, Saito M, Tsukada J, et al: Vasopressin increases

ascular endothelial growth factor secretion from human vascular

mooth muscle cells. Eur J Pharmacol 368:89-94, 1999

Ya1

bJ

nav

ct

eo2

esm5

eot

itf2

ct3

P

aJ

mH

p1

dp

b2

ns

pl

aiJ

aaS

apd

pt

H

pp

ab

it

pd1

ar2

8s8

rs

ob

dr

dS

sa

mS

hC

cs

pt

w

343VASOPRESSIN

99. Tahara A, Tomura Y, Wada K, et al: Pharmacological profile ofM087, a novel potent nonpeptide vasopressin V1A and V2 receptor

ntagonist, in vitro and in vivo. J Pharmacol Exp Ther 282:301-308,997100. Tahara A, Tsukada J, Ishii N, et al: Comparison of vasopressin

inding sites in human uterine and vascular smooth muscle cells. EurPharmacol 378:137-142, 1999101. Tahara A, Tomura Y, Wada K, et al: Effect of YM087, a potent

onpeptide vasopressin antagonist, on vasopressin-induced hyperplasiand hypertrophy of cultured vascular smooth-muscle cells. J Cardio-asc Pharmacol 30:759-766, 1997102. Nakamura Y, Haneda T, Osaki J, et al: Hypertrophic growth of

ultured neonatal rat heart cells mediated by vasopressin V(1A) recep-or. Eur J Pharmacol 391:39-48, 2000

103. Udelson J, Smith W, Hendrix G, et al: Acute hemodynamicffects of Conivaptan, a dual V1a and V2 vasopressin receptor antag-nist, in patients with advanced heart failure. Circulation 104:2417-423, 2001104. Udelson J, Orlandi C, Ouyang J, et al: Acute hemodynamic

ffects of tolvaptan, a vasopressin V2 receptor blocker, in patients withymptomatic heart failure and systolic dysfunction: an international,ulticenter, randomized, placebo-controlled trial. J Am Coll Cardiol

2:1540-1545, 2008105. Russell SD, Selaru P, Pyne DA, et al: Rationale for use of an

xercise end point and design for the ADVANCE (A Dose evaluationf a Vasopressin ANtagonist in CHF patients undergoing Exercise)rial. Am Heart J 145:179-186, 2003

106. Udelson J, McGrew F, Flores E, et al: Multicenter, random-zed, double-blind, placebo-controlled study on the effect of oralolvaptan on left ventricular dilation and function in patients with heartailure and systolic dysfunction. J Am Coll Cardiol 49:2151-2159,007107. Bichet D, Razi M, Lonergan M, et al: Hemodynamic and

oagulation responses to 1-desamino[8-D-arginine] vasopressin in pa-ients with congenital nephrogenic diabetes insipidus. N Engl J Med18:881-887, 1988108. Robertson J, Lillicrap D, James P: von Willebrand Disease.

ediatr Clin North Am 55:377-392, 2008109. Mannucci P, Remuzzi G, Pusineri F, et al: Deamino-8-D-

rginine vasopressin shortens the bleeding time in uremia. N EnglMed 308:8-12, 1983110. Viganò G, Mannucci P, Lattuada A, et al: Subcutaneous des-opressin (DDAVP) shortens the bleeding time in uremia. Am Jematol 31:32-35, 1989111. Shapiro M, Kelleher S: Intranasal deamino-8-D-arginine vaso-

ressin shortens the bleeding time in uremia. Am J Nephrol 4:260-261,984112. Mannucci PM, Vicente V, Vianello L, et al: Controlled trial of

esmopressin in liver cirrhosis and other conditions associated with arolonged bleeding time. Blood 67:1148-1153, 1986113. Burroughs A, Matthews K, Qadiri M, et al: Desmopressin and

leeding time in patients with cirrhosis. Br Med J (Clin Res Ed)91:1377-1381, 1985114. López P, Otaso J, Alvarez D, et al: Hemostatic and hemody-

amic effects of vasopressin analogue DDAVP in patients with cirrho-is. Acta Gastroenterol Latinoam 27:59-62, 1997

115. Cattaneo M, Tenconi P, Alberca I, et al: Subcutaneous desmo-ressin (DDAVP) shortens the prolonged bleeding time in patients withiver cirrhosis. Thromb Haemost 64:358-360, 1990

116. Sheridan D, Card R, Pinilla J, et al: Use of desmopressincetate to reduce blood transfusion requirements during cardiac surgeryn patients with acetylsalicylic-acid-induced platelet dysfunction. Can

Surg 37:33-36, 1994 p

117. Gratz I, Koehler J, Olsen D, et al: The effect of desmopressincetate on postoperative hemorrhage in patients receiving aspirin ther-py before coronary artery bypass operations. J Thorac Cardiovascurg 104:1417-1422, 1992118. Kobrinsky NL, Letts RM, Patel LR, et al: 1-Desamino-8-D-

rginine vasopressin (desmopressin) decreases operative blood loss inatients having Harrington rod spinal fusion surgery. A randomized,ouble-blinded, controlled trial. Ann Intern Med 107:446-450, 1987119. Cattaneo M, Lombardi R, Bettega D, et al: Shear-induced

latelet aggregation is potentiated by desmopressin and inhibited byiclopidine. Arterioscler Thromb 13:393-397, 1993

120. Cattaneo M: The use of desmopressin in open-heart surgery.aemophilia 14:40-47, 2008 (suppl 1)121. Flordal P, Ljungström K, Ekman B, et al: Effects of desmo-

ressin on blood loss in hip arthroplasty. Controlled study in 50atients. Acta Orthop Scand 63:381-385, 1992122. Rocha E, Llorens R, Paramo JA, et al: Does desmopressin

cetate reduce blood loss after surgery in patients on cardiopulmonaryypass? Circulation 77:1319-1323, 1988123. Hackmann T, Naiman SC: Con: Desmopressin is not of value

n the treatment of post-cardiopulmonary bypass bleeding. J Cardio-horac Vasc Anesth 5:290-293, 1991

124. Ansell J, Klassen V, Lew R, et al: Does desmopressin acetaterophylaxis reduce blood loss after valvular heart operations? A ran-omized, double-blind study. J Thorac Cardiovasc Surg 104:117-123,992125. Theroux M, Corddry D, Tietz A, et al: A study of desmopressin

nd blood loss during spinal fusion for neuromuscular scoliosis: Aandomized, controlled, double-blinded study. Anesthesiology 87:260-67, 1997126. Alanay A, Acaroglu E, Ozdemir O, et al: Effects of deamino-

-D-arginine vasopressin on blood loss and coagulation factors incoliosis surgery. A double-blind randomized clinical trial. Spine 24:77-882, 1999127. Guay J, Reinberg C, Poitras B, et al: A trial of desmopressin to

educe blood loss in patients undergoing spinal fusion for idiopathiccoliosis. Anesth Analg 75:405-410, 1992

128. Karnezis T, Stulberg S, Wixson R, et al: The hemostatic effectsf desmopressin on patients who had total joint arthroplasty. A double-lind randomized trial. J Bone Joint Surg Am 76:1545-1550, 1994129. Schött U, Sollén C, Axelsson K, et al: Desmopressin acetate

oes not reduce blood loss during total hip replacement in patientseceiving dextran. Acta Anaesthesiol Scand 39:592-598, 1995

130. Fremes S, Wong B, Lee E, et al: Metaanalysis of prophylacticrug treatment in the prevention of postoperative bleeding. Ann Thoracurg 58:1580-1588, 1994131. Levi M, Cromheecke M, de Jonge E, et al: Pharmacological

trategies to decrease excessive blood loss in cardiac surgery: A meta-nalysis of clinically relevant endpoints. Lancet 354:1940-1947, 1999

132. Carless P, Henry D, Moxey A, et al: Desmopressin for mini-ising perioperative allogeneic blood transfusion. Cochrane Databaseyst Rev CD001884, 2004133. Döhler K, Meyer M: Vasopressin analogues in the treatment of

epatorenal syndrome and gastrointestinal haemorrhage. Best Pract Reslin Anaesthesiol 22:335-350, 2008134. Testo A, Wongseelashote S, Angus P, et al: Long-term out-

ome of patients treated with terlipressin for types 1 and 2 hepatorenalyndrome. J Gastroenterol Hepatol 23:1535-1540, 2007

135. Sanyal A, Boyer T, Garcia-Tsao G, et al: A randomized,rospective, double-blind, placebo-controlled trial of terlipressin forype 1 hepatorenal syndrome. Gastroenterology 134:1360-1368, 2008

136. Ortega R, Ginès P, Uriz J, et al: Terlipressin therapy with andithout albumin for patients with hepatorenal syndrome: Results of a

rospective, nonrandomized study. Hepatology 36:941-948, 2002

wt

aD

tc

c1

eo

s

c2

r

s

h1

ps

os

N

vS

ctt

dO

c

a3

e3

iC

r

c

sp

ns

o2

it

c

iP

c

1

ps

v7

mt

ir

di

vd

isc

vp

pp2

np3

r

Cl

vs

e

344 HOLT AND HASPEL

137. Moreau R, Durand F, Poynard T, et al: Terlipressin in patientsith cirrhosis and type 1 hepatorenal syndrome: A retrospective mul-

icenter study. Gastroenterology 122:923-930, 2002138. Neri S, Pulvirenti D, Malaguarnera M, et al: Terlipressin and

lbumin in patients with cirrhosis and type I hepatorenal syndrome. Digis Sci 53:830-835, 2008139. Solanki P, Chawla A, Garg R, et al: Beneficial effects of

erlipressin in hepatorenal syndrome: A prospective, randomized pla-ebo-controlled clinical trial. J Gastroenterol Hepatol 18:152-156, 2003

140. Landry D, Levin H, Gallant E, et al: Vasopressin deficiencyontributes to the vasodilation of septic shock. Circulation 95:1122-125, 1997141. O’Brien A, Wilson A, Sibbald R, et al: Temporal variation in

ndotoxin-induced vascular hyporeactivity in a rat mesenteric arteryrgan culture model. Br J Pharmacol 133:351-360, 2001142. Buckley J, Singer M, Clapp L: Role of KATP channels in

epsis. Cardiovasc Res 72:220-230, 2006143. Sharshar T, Annane D: Endocrine effects of vasopressin in

ritically ill patients. Best Pract Res Clin Anaesthesiol 22:265-273,008144. Lin I, Ma H, Lin A, et al: Low plasma vasopressin/norepineph-

ine ratio predicts septic shock. Am J Emerg Med 23:718-724, 2005145. Reid I: Role of vasopressin deficiency in the vasodilation of

eptic shock. Circulation 95:1108-1110, 1997146. Landry DW, Levin HR, Gallant EM, et al: Vasopressin pressor

ypersensitivity in vasodilatory septic shock. Crit Care Med 25:1279-282, 1997147. Morales D, Madigan J, Cullinane S, et al: Reversal by vaso-

ressin of intractable hypotension in the late phase of hemorrhagichock. Circulation 100:226-229, 1999

148. Errington ML, Rocha e Silva MJ: The secretion and clearancef vasopressin during the development of irreversible haemorrhagichock. J Physiol 217:43P-45P, 1971

149. Landry D, Oliver J: The pathogenesis of vasodilatory shock.Engl J Med 345:588-595, 2001150. Wilson M, Brackett D, Tompkins P, et al: Elevated plasma

asopressin concentrations during endotoxin and E. coli shock. Advhock Res 6:15-26, 1981151. Brackett D, Schaefer C, Tompkins P, et al: Evaluation of

ardiac output, total peripheral vascular resistance, and plasma concen-rations of vasopressin in the conscious, unrestrained rat during endo-oxemia. Circ Shock 17:273-284, 1985

152. Wilson M, Brackett D, Hinshaw L, et al: Vasopressin releaseuring sepsis and septic shock in baboons and dogs. Surg Gynecolbstet 153:869-872, 1981153. Wilson M, Brackett D: Release of vasoactive hormones and

irculatory changes in shock. Circ Shock 11:225-234, 1983154. Barrett LK, Singer M, Clapp LH: Vasopressin: Mechanisms of

ction on the vasculature in health and in septic shock. Crit Care Med5:33-40, 2007155. Russell J, Walley K, Singer J, et al: Vasopressin versus nor-

pinephrine infusion in patients with septic shock. N Engl J Med58:877-887, 2008156. Russell J, Walley K, Gordon A, et al: Interaction of vasopressin

nfusion, corticosteroid treatment, and mortality of septic shock. Critare Med 37:811-818, 2009157. O’Brien A, Clapp L, Singer M: Terlipressin for norepineph-

ine-resistant septic shock. Lancet 359:1209-1210, 2002158. Leone M, Albanese J, Delmas A, et al: Terlipressin in cate-

holamine-resistant septic shock patients. Shock 22:314-319, 2004159. Morelli A, Rocco M, Conti G, et al: Effects of terlipressin on

ystemic and regional haemodynamics in catecholamine-treated hy-

erkinetic septic shock. Intensive Care Med 30:597-604, 2004 N

160. Albanèse J, Leone M, Delmas A, et al: Terlipressin or norepi-ephrine in hyperdynamic septic shock: A prospective, randomizedtudy. Crit Care Med 33:1897-1902, 2005

161. Morelli A, Ertmer C, Westphal M: Terlipressin in the treatmentf septic shock: The earlier the better? Best Pract Res Clin Anesthesiol2:317-321, 2008162. Morelli A, Ertmer C, Lange M, et al: Continuous terlipressin

nfusion in patients with septic shock: Less may be best, and the earlierhe better? Intensive Care Med 33:1669-1670, 2007

163. Meyer S, Gortner L, McGuire W, et al: Vasopressin in cate-holamine-refractory shock in children. Anaesthesia 63:228-234, 2008

164. Leone M, Martin C: Role of terlipressin in the treatment ofnfants and neonates with catecholamine-resistant septic shock. Bestract Res Clin Anesthesiol 22:323-333, 2008165. Choong K, Kissoon N: Vasopressin in pediatric shock and

ardiac arrest. Pediatr Crit Care Med 9:372-379, 2008166. Wolf A: Vasopressin in paediatric practice. Paediatr Anaesth

8:579-581, 2008167. Dellinger R, Levy M, Carlet J, et al: Surviving Sepsis Cam-

aign: International guidelines for management of severe sepsis andeptic shock: 2008. Intensive Care Med 34:17-60, 2008

168. Lindner KH, Haak T, Keller A, et al: Release of endogenousasopressors during and after cardiopulmonary resuscitation. Heart5:145-150, 1996169. Lindner KH, Strohmenger HU, Ensinger H, et al: Stress hor-one response during and after cardiopulmonary resuscitation. Anes-

hesiology 77:662-668, 1992170. Lindner KH, Prengel AW, Pfenninger EG, et al: Vasopressin

mproves vital organ blood flow during closed-chest cardiopulmonaryesuscitation in pigs. Circulation 91:215-221, 1995

171. Prengel AW, Lindner KH, Keller A: Cerebral oxygenationuring cardiopulmonary resuscitation with epinephrine and vasopressinn pigs. Stroke 27:1241-1248, 1996

172. Wenzel V, Lindner KH, Augenstein S, et al: Intraosseousasopressin improves coronary perfusion pressure rapidly during car-iopulmonary resuscitation in pigs. Crit Care Med 27:1565-1569, 1999173. Wenzel V, Lindner KH, Krismer AC, et al: Repeated admin-

stration of vasopressin but not epinephrine maintains coronary perfu-ion pressure after early and late administration during prolongedardiopulmonary resuscitation in pigs. Circulation 99:1379-1384, 1999

174. Wenzel V, Lindner KH, Prengel AW, et al: Endobronchialasopressin improves survival during cardiopulmonary resuscitation inigs. Anesthesiology 86:1375-1381, 1997175. Wenzel V, Lindner KH, Prengel AW, et al: Vasopressin im-

roves vital organ blood flow after prolonged cardiac arrest withostcountershock pulseless electrical activity in pigs. Crit Care Med7:486-492, 1999176. Wenzel V, Lindner KH, Krismer AC, et al: Survival with full

eurologic recovery and no cerebral pathology after prolonged cardio-ulmonary resuscitation with vasopressin in pigs. J Am Coll Cardiol5:527-533, 2000177. Aung K, Htay T: Vasopressin for cardiac arrest: A systematic

eview and meta-analysis. Arch Int Med 165:17-24, 2005178. Nolan J, Deakin C, Soar J, et al: European Resuscitation

ouncil guidelines for resuscitation 2005. Section 4. Adult advancedife support. Resuscitation 67:S39-S86, 2005 (suppl 1)

179. Wenzel V, Krismer AC, Arntz HR, et al: A comparison ofasopressin and epinephrine for out-of-hospital cardiopulmonary re-uscitation. N Engl J Med 350:105-113, 2004

180. Gueugniaud P-Y, David J-S, Chanzy E, et al: Vasopressin andpinephrine vs. epinephrine alone in cardiopulmonary resuscitation.

Engl J Med 359:21-30, 2008

pA

s5

hAd

m1

iA

sr2

pmp

A

srt

cv5

oo

ps

oA

sC

vB

es1

ba

i3

Ag

[

o5

nai

ti

ntt

riis

etA

rc

v6

t1

vf1

dJ

Poa

d5

p2

c

aa7

vS

ds2

f

345VASOPRESSIN

181. Mentzelopoulos SD, Zakynthinos SG, Tzoufi M, et al: Vaso-ressin, epinephrine, and corticosteroids for in-hospital cardiac arrest.rch Int Med 169:15-24, 2009182. Mann K, Berg RA, Nadkarni V: Beneficial effects of vasopres-

in in prolonged pediatric cardiac arrest: A case series. Resuscitation2:149-156, 2002183. Duncan J, Meaney P, Simpson P, et al: Vasopressin for in-

ospital pediatric cardiac arrest: Results from the American Heartssociation National Registry of Cardiopulmonary Resuscitation. Pe-iatr Crit Care Med 10:191-195, 2009184. 2005 American Heart Association Guidelines for Cardiopul-onary Resuscitation and Emergency Cardiovascular Care. Circulation

12:IV1-203, 2005185. Voelckel WG, Lurie KG, Lindner KH, et al: Vasopressin

mproves survival after cardiac arrest in hypovolemic shock. Anesthnalg 91:627-634, 2000186. Voelckel WG, Raedler C, Wenzel V, et al: Arginine vasopres-

in, but not epinephrine, improves survival in uncontrolled hemor-hagic shock after liver trauma in pigs. Crit Care Med 31:1160-1165,003187. Stadlbauer KH, Wagner-Berger HG, Raedler C, et al: Vaso-

ressin, but not fluid resuscitation, enhances survival in a liver traumaodel with uncontrolled and otherwise lethal hemorrhagic shock in

igs. Anesthesiology 98:699-704, 2003188. Sharma RM, Setlur R: Vasopressin in hemorrhagic shock.

nesth Analg 101:833-834, 2005189. Krismer AC, Wenzel V, Voelckel WG, et al: Employing va-

opressin as an adjunct vasopressor in uncontrolled traumatic hemor-hagic shock. Three cases and a brief analysis of the literature. Anaes-hesist 54:220-224, 2005

190. Haas T, Voelckel W, Wiedermann F, et al: Successful resus-itation of a traumatic cardiac arrest victim in hemorrhagic shock withasopressin: A case report and brief review of the literature. J Trauma7:177-179, 2004191. Johnson K, Pearce F, Jeffreys N, et al: Impact of vasopressin

n hemodynamic and metabolic function in the decompensatory phasef hemorrhagic shock. J Cardiothorac Vasc Anesth 20:167-172, 2006192. Wenzel V, Raab H, Dunser MW: Arginine vasopressin: A

romising rescue drug in the treatment of uncontrolled haemorrhagichock. Best Pract Res Clin Anaesthesiol 22:299-316, 2008

193. Lienhart H, Wenzel V, Braun J, et al: [Vasopressin for therapyf persistent traumatic hemorrhagic shock: The VITRIS.at study].naesthesist 56:145-148, 2007194. Lienhart HG, Lindner KH, Wenzel V: Developing alternative

trategies for the treatment of traumatic haemorrhagic shock. Curr Opinrit Care 14:247-253, 2008195. Jochberger S, Dunser MW: Arginine vasopressin as a rescue

asopressor to treat epidural anaesthesia-induced arterial hypotension.est Pract Res Clin Anaesthesiol 22:383-391, 2008196. Hopf H, Arand D, Peters J: Sympathetic blockade by thoracic

pidural anaesthesia suppresses renin release in response to hypoten-ion, but activates the vasopressin system. Eur J Anaesthesiol 9:63-69,992197. Hopf H, Schlaghecke R, Peters J: Sympathetic neural blockade

y thoracic epidural anesthesia suppresses renin release in response torterial hypotension. Anesthesiology 80:992-999, 1994

198. Lange M, Ertmer C, Westphal M: Vasopressin vs. terlipressinn the treatment of cardiovascular failure in sepsis. Intensive Care Med4:821-832, 2008199. De Kock M, Laterre P, Andruetto P, et al: Ornipressin (Por 8):

n efficient alternative to counteract hypotension during combinedeneral/epidural anesthesia. Anesth Analg 90:1301-1307, 2000200. Vaquero Roncero L, Sánchez Montero F, Muriel Villoria C:

Effectiveness of epidural administration of saline solutions to prevent S

r treat postdural puncture headache]. Rev Esp Anestesiol Reanim1:589-594, 2004201. Boccara G, Ouattara A, Godet G, et al: Terlipressin versus

orepinephrine to correct refractory arterial hypotension after generalnesthesia in patients chronically treated with renin-angiotensin systemnhibitors. Anesthesiology 98:1338-1344, 2003

202. Braun E, Palin C, Hogue C: Vasopressin during spinal anes-hesia in a patient with primary pulmonary hypertension treated withntravenous epoprostenol. Anesth Analg 99:36-37, 2004

203. Morelli A, Tritapepe L, Rocco M, et al: Terlipressin versusorepinephrine to counteract anesthesia-induced hypotension in pa-ients treated with renin-angiotensin system inhibitors: Effects on sys-emic and regional hemodynamics. Anesthesiology 102:12-19, 2005

204. Meersschaert K, Brun L, Gourdin M, et al: Terlipressin-ephed-ine versus ephedrine to treat hypotension at the induction of anesthesian patients chronically treated with angiotensin-converting enzymenhibitors: A prospective, randomized, double-blinded, crossovertudy. Anesth Analg 94:835-840, 2002

205. Eyraud D, Brabant S, Nathalie D, et al: Treatment of intraop-rative refractory hypotension with terlipressin in patients chronicallyreated with an antagonist of the renin-angiotensin system. Anesthnalg 88:980-984, 1999206. Morales D, Garrido M, Madigan J, et al: A double-blind

andomized trial: prophylactic vasopressin reduces hypotension afterardiopulmonary bypass. Ann Thorac Surg 75:926-930, 2003

207. Schummer C, Wirsing M, Schummer W: The pivotal role ofasopressin in refractory anaphylactic shock. Anesth Analg 107:620-24, 2008208. Schummer W, Schummer C, Wippermann J, et al: Anaphylac-

ic shock: Is vasopressin the drug of choice? Anesthesiology 101:1025-027, 2004209. Argenziano M, Chen JM, Choudhri AF, et al: Management of

asodilatory shock after cardiac surgery: Identification of predisposingactors and use of a novel pressor agent. J Thorac Cardiovasc Surg16:973-980, 1998210. Paparella D, Yau T, Young E: Cardiopulmonary bypass in-

uced inflammation: Pathophysiology and treatment. An update. EurCardiothorac Surg 21:232-244, 2002211. Hoshikawa-Fujimura A, Auler Júnior J, Da Rocha T, et al:

AF-acether, superoxide anion and beta-glucuronidase as parametersf polymorphonuclear cell activation associated with cardiac surgerynd cardiopulmonary bypass. Braz J Med Biol Res 22:1077-1082, 1989

212. Kirklin J: Prospects for understanding and eliminating theeleterious effects of cardiopulmonary bypass. Ann Thorac Surg 51:29-531, 1991213. Singh Ranger G: Antidiuretic hormone replacement therapy to

revent or ameliorate vasodilatory shock. Med Hypotheses 59:337-340,002214. Wu W, Zbuzek VK, Bellevue C: Vasopressin release during

ardiac operation. J Thorac Cardiovasc Surg 79:83-90, 1980215. Kuitunen A, Hynynen M, Salmenpera M, et al: Anaesthesia

ffects plasma concentrations of vasopressin, von Willebrand factornd coagulation factor VIII in cardiac surgical patients. Br J Anaesth0:173-180, 1993216. Woods WG, Forsling ML, Le Quesne LP: Plasma arginine

asopressin levels and arterial pressure during open heart surgery. Br Jurg 76:29-32, 1989217. Argenziano M, Choudhri A, Oz M, et al: A prospective ran-

omized trial of arginine vasopressin in the treatment of vasodilatoryhock after left ventricular assist device placement. Circulation 96:II-86-II-290, 1997218. Masetti P, Murphy SF, Kouchoukos NT: Vasopressin therapy

or vasoplegic syndrome following cardiopulmonary bypass. J Cardiac

urg 17:485-489, 2002

dM

pa9

t1

ll

pM

eI

e1

iA

sl

t1

as

ii2

ims

Im

ia8

Kc

sa

p7

c2

pr

v

vt

omG

ran

4n

Sp

a1

C2

v

tt

da

K

P

ip

e

N

e

Si1

viB

tn

vs2

346 HOLT AND HASPEL

219. Dunser MW, Mayr AJ, Stallinger A, et al: Cardiac performanceuring vasopressin infusion in postcardiotomy shock. Intensive Careed 28:746-751, 2002220. Dunser MW, Mayr AJ, Ulmer H, et al: The effects of vaso-

ressin on systemic hemodynamics in catecholamine-resistant septicnd postcardiotomy shock: A retrospective analysis. Anesth Analg3:7-13, 2001221. Overand PT, Teply JF: Vasopressin for the treatment of refrac-

ory hypotension after cardiopulmonary bypass. Anesth Analg 86:207-1209, 1998222. Mayr VD, Wenzel V, Muller T, et al: Effects of vasopressin on

eft anterior descending coronary artery blood flow during extremelyow cardiac output. Resuscitation 62:229-235, 2004

223. Luckner G, Dunser MW, Jochberger S, et al: Arginine vaso-ressin in 316 patients with advanced vasodilatory shock. Crit Careed 33:2659-2666, 2005224. Shelly MP, Greatorex R, Calne RY, et al: The physiological

ffects of vasopressin when used to control intra-abdominal bleeding.ntensive Care Med 14:526-531, 1988

225. Sirinek KR, Thomford NR: Isoproterenol in offsetting adverseffects of vasopressin in cirrhotic patients. Am J Surg 129:130-136,975226. Morales DL, Gregg D, Helman DN, et al: Arginine vasopressin

n the treatment of 50 patients with postcardiotomy vasodilatory shock.nn Thorac Surg 69:102-106, 2000227. Egi M, Bellomo R, Langenberg C, et al: Selecting a vasopres-

or drug for vasoplegic shock after adult cardiac surgery: A systematiciterature review. Ann Thorac Surg 83:715-723, 2007

228. Leone M, Charvet A, Delmas A, et al: Terlipressin: A newherapeutic for calcium-channel blockers overdose. J Crit Care 20:114-15, 2005229. Zuidema X, Dünser M, Wenzel V, et al: Terlipressin as an

djunct vasopressor in refractory hypotension after tricyclic antidepres-ant intoxication. Resuscitation 72:319-323, 2007

230. Salluh J, Martins G, Santino M, et al: Early use of terlipressinn catecholamine-resistant shock improves cerebral perfusion pressuren severe traumatic brain injury. Acta Anaesthesiol Scand 51:505-508,007231. Kusano E, Tian S, Umino T, et al: Arginine vasopressin inhib-

ts interleukin-1 beta-stimulated nitric oxide and cyclic guanosineonophosphate production via the V1 receptor in cultured rat vascular

mooth muscle cells. J Hypertens 15:627-632, 1997232. Umino T, Kusano E, Muto S, et al: AVP inhibits LPS- and

L-1beta-stimulated NO and cGMP via V1 receptor in cultured ratesangial cells. Am J Physiol 276:F433-441, 1999233. Nambi P, Whitman M, Gessner G, et al: Vasopressin-mediated

nhibition of atrial natriuretic factor-stimulated cGMP accumulation inn established smooth muscle cell line. Proc Natl Acad Sci U S A3:8492-8495, 1986234. Wakatsuki T, Nakaya Y, Inoue I: Vasopressin modulates

(�)-channel activities of cultured smooth muscle cells from porcineoronary artery. Am J Physiol 263:H491-H496, 1992

235. Wakatsuki T, Nakaya Y, Miyoshi Y, et al: Effects of vasopres-in on ATP-sensitive and Ca(2�)-activated K� channels of coronaryrterial smooth muscle cells. Jpn J Pharmacol 58:339P, 1992 (suppl 2)

236. Malay MB, Ashton RC Jr, Landry DW, et al: Low-dose vaso-ressin in the treatment of vasodilatory septic shock. J Trauma 47:699-03, 1999237. Bartelstone H, Nasmyth P: Vasopressin potentiation of cate-

holamine actions in dog, rat, cat, and rat aortic strip. Am J Physiol08:754-762, 1965238. Medina P, Acuña A, Martínez-León J, et al: Arginine vaso-

ressin enhances sympathetic constriction through the V1 vasopressin

eceptor in human saphenous vein. Circulation 97:865-870, 1998 n

239. Buijk S, Bruining H: Vasopressin deficiency contributes to theasodilation of septic shock. Circulation 98:187, 1998240. Hayoz D, Bizzini G, Noel B, et al: Effect of SR 49059, a V1a

asopressin receptor antagonist, in Raynaud’s phenomenon. Rheuma-ology 39:1132-1138, 2000

241. Akerlund M, Bossmar T, Brouard R, et al: Receptor binding ofxytocin and vasopressin antagonists and inhibitory effects on isolatedyometrium from preterm and term pregnant women. Br J Obstetynaecol 106:1047-1053, 1999242. Steinwall M, Bossmar T, Brouard R, et al: The effect of

elcovaptan (SR 49059), an orally active vasopressin V1a receptorntagonist, on uterine contractions in preterm labor. Gynecol Endocri-ol 20:104-109, 2005243. Steinwall M, Bossmar T, Gaud C, et al: Inhibitory effects of SR

9059 on oxytocin and vasopressin-induced uterine contractions inon-pregnant women. Acta Obstet Gynecol Scand 83:12-18, 2004244. Brouard R, Bossmar T, Fournie-Lloret D, et al: Effect of

R49059, an orally active V1a vasopressin receptor antagonist, in therevention of dysmenorrhoea. BJOG 107:614-619, 2000245. Takeda T, Sawada S, Takeda S, et al: The effects of V2

ntagonist (OPC-31260) on endolymphatic hydrops. Hear Res 182:9-8, 2003246. Sonino N, Boscaro M, Fallo F: Pharmacologic management of

ushing syndrome: New targets for therapy. Treat Endocrinol 4:87-94,005247. Ali F, Guglin M, Vaitkevicius P, et al: Therapeutic potential of

asopressin receptor antagonists. Drugs 67:847-858, 2007248. Doggrell S: Do vasopressin receptor type 2 antagonists have

herapeutic potential in polycystic kidney diseases? Expert Opin Inves-ig Drugs 13:431-434, 2004

249. Gattone VN, Wang X, Harris P, et al: Inhibition of renal cysticisease development and progression by a vasopressin V2 receptorntagonist. Nat Med 9:1323-1326, 2003

250. Greenberg A, Verbalis JG: Vasopressin receptor antagonists.idney Int 69:2124-2130, 2006251. Gross P: Polycystic kidney disease: Will it become treatable?

ol Arch Med Wewn 118:298-301, 2008252. Mastorakos G, Pavlatou MG, Mizamtsidi M: The hypothalam-

c-pituitary-adrenal and the hypothalamic- pituitary-gonadal axes inter-lay. Pediatr Endocrinol Rev 3:172-181, 2006 (suppl 1)253. Torres VE: Vasopressin antagonists in polycystic kidney dis-

ase. Kidney Int 68:2405-2418, 2005254. Torres VE: Role of vasopressin antagonists. Clin J Am Soc

ephrol 3:1212-1218, 2008255. Torres VE: Vasopressin antagonists in polycystic kidney dis-

ase. Sem Nephrol 28:306-317, 2008256. Lacheretz F, Barbier A, Serradeil-Le Gal C, et al: Effect of

R121463, a selective non-peptide vasopressin V2 receptor antagonist,n a rabbit model of ocular hypertension. J Ocul Pharmacol Ther6:203-216, 2000257. Naito A, Kurasawa T, Ohtake Y, et al: The effects of several

asopressin receptor antagonists on normal intraocular pressure and thentraocular distribution of vasopressin receptor subtypes. Biol Pharmull 25:251-255, 2002258. Nishihashi T, Trandafir C, Wang A, et al: Enhanced reactivity

o vasopressin in rat basilar arteries during vasospasm after subarach-oid hemorrhage. Eur J Pharmacol 513:93-100, 2005259. Trandafir C, Nishihashi T, Wang A, et al: Participation of

asopressin in the development of cerebral vasospasm in a rat model ofubarachnoid haemorrhage. Clin Exp Pharmacol Physiol 31:261-266,004260. Rabinstein AA: Vasopressin antagonism: Potential impact on

eurologic disease. Clin Neuropharmacol 29:87-93, 2006

bO

h2

E3

Vr8

c2

s2

av2

aC

d

r1

am

art9

rT

sm2

vs

a

a2

aa1

s

fC

cr3

vs

l3

as

ep

so

i3

S

aA

v3

os2

ps

t

347VASOPRESSIN

261. Molnár A, Varga C, Berkó A, et al: Prevention of hypoxicrain oedema by the administration of vasopressin receptor antagonistPC-31260. Prog Brain Res 170:519-525, 2008262. Caldwell HK, Lee H-J, Macbeth AH, et al: Vasopressin: Be-

avioral roles of an “original” neuropeptide. Prog Neurobiol 84:1-24,008263. Heinrichs M, Domes G: Neuropeptides and social behaviour:

ffects of oxytocin and vasopressin in humans. Prog Brain Res 170:37-350, 2008264. Hodgson RA, Higgins GA, Guthrie DH, et al: Comparison of the

1b antagonist, SSR149415, and the CRF1 antagonist, CP-154,526, inodent models of anxiety and depression. Pharmacol Biochem Behav6:431-440, 2007

265. Keck ME: Corticotropin-releasing factor, vasopressin and re-eptor systems in depression and anxiety. Amino Acids 31:241-250,006266. Landgraf R: The involvement of the vasopressin system in

tress-related disorders. CNS Neurol Disord Drug Targets 5:167-179,006267. Landgraf R, Kessler MS, Bunck M, et al: Candidate genes of

nxiety-related behavior in HAB/LAB rats and mice: Focus onasopressin and glyoxalase-I. Neurosci Biobehav Rev 31:89-102,007268. Simon NG, Guillon C, Fabio K, et al: Vasopressin antagonists

s anxiolytics and antidepressants: recent developments. Recent PatNS Drug Discov 3:77-93, 2008269. Surget A, Belzung C: Involvement of vasopressin in affective

isorders. Eur J Pharmacol 583:340-349, 2008270. Veenema AH, Neumann ID: Central vasopressin and oxytocin

elease: Regulation of complex social behaviours. Prog Brain Res70:261-276, 2008271. Blanchard RJ, Griebel G, Farrokhi C, et al: AVP V1b selective

ntagonist SSR149415 blocks aggressive behaviors in hamsters. Phar-acol Biochem Behav 80:189-194, 2005272. Griebel G, Simiand J, Serradeil-Le Gal C, et al: Anxiolytic-

nd antidepressant-like effects of the non-peptide vasopressin V1beceptor antagonist, SSR149415, suggest an innovative approach forhe treatment of stress-related disorders. Proc Natl Acad Sci U S A9:6370-6375, 2002273. Griebel G, Simiand J, Stemmelin J, et al: The vasopressin V1b

eceptor as a therapeutic target in stress-related disorders. Curr Drugargets CNS Neurol Disord 2:191-200, 2003274. Griebel G, Stemmelin J, Gal CS-L, et al: Non-peptide va-

opressin V1b receptor antagonists as potential drugs for the treat-ent of stress-related disorders. Curr Pharm Des 11:1549-1559,

005275. Overstreet DH, Griebel G: Antidepressant-like effects of the

asopressin V1b receptor antagonist SSR149415 in the Flinders Sen-itive Line rat. Pharm Biochem Behav 82:223-227, 2005

276. Serradeil-Le Gal C, Derick S, Brossard G, et al: Functional

nd pharmacological characterization of the first specific agonist 2

nd antagonist for the V1b receptor in mammals. Stress 6:199-206,003277. Dempster EL, Burcescu I, Wigg K, et al: Evidence of an

ssociation between the vasopressin V1b receptor gene (AVPR1B)nd childhood-onset mood disorders. Arch Gen Psych 64:1189-195, 2007278. Cartotto R, McGibney K, Smith T, et al: Vasopressin for the

eptic burn patient. Burns 33:441-451, 2007279. Bunker N, Higgins D: Peripheral administration of vasopressin

or catecholamine-resistant hypotension complicated by skin necrosis.rit Care Med 34:935, 2006280. Dünser M, Mayr A, Tür A, et al: Ischemic skin lesions as a

omplication of continuous vasopressin infusion in catecholamine-esistant vasodilatory shock: Incidence and risk factors. Crit Care Med1:1394-1398, 2003281. Kim E, Lee S, Byun S, et al: Skin necrosis after a low-dose

asopressin infusion through a central venous catheter for treatingeptic shock. Korean J Intern Med 21:287-290, 2006

282. Kahn J, Kress J, Hall J: Skin necrosis after extravasation ofow-dose vasopressin administered for septic shock. Crit Care Med0:1899-1901, 2002283. Dunser MW, Mayr AJ, Ulmer H, et al: Arginine vasopressin in

dvanced vasodilatory shock: A prospective, randomized, controlledtudy. Circulation 107:2313-2319, 2003

284. van Haren FMP, Rozendaal FW, van der Hoeven JG: Theffect of vasopressin on gastric perfusion in catecholamine-dependentatients in septic shock. Chest 124:2256-2260, 2003285. Patel BM, Chittock DR, Russell JA, et al: Beneficial effects of

hort-term vasopressin infusion during severe septic shock. Anesthesi-logy 96:576-582, 2002286. Klinzing S, Simon M, Reinhart K, et al: High-dose vasopressin

s not superior to norepinephrine in septic shock. Crit Care Med1:2646-2650, 2003287. Russell JA: Vasopressin in septic shock. Crit Care Med 35:

609-615, 2007288. Asfar P, Bracht H, Radermacher P: Impact of vasopressin

nalogues on the gut mucosal microcirculation. Best Pract Res Clinnaesthesiol 22:351-358, 2008289. Obritsch MD, Jung R, Fish DN, et al: Effects of continuous

asopressin infusion in patients with septic shock. Ann Pharmacother8:1117-1122, 2004290. Holmes CL, Walley KR, Chittock DR, et al: The effects

f vasopressin on hemodynamics and renal function in severeeptic shock: A case series. Intensive Care Med 27:1416-1421,001291. Dünser M, Fries D, Schobersberger W, et al: Does arginine vaso-

ressin influence the coagulation system in advanced vasodilatory shock withevere multiorgan dysfunction syndrome? Anesth Analg 99:201-206, 2004

292. Dunser MW, Westphal M: Arginine vasopressin in vasodila-ory shock: Effects on metabolism and beyond. Curr Opin Anaesthesiol

1:122-127, 2008