The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may...

18
THE PRESENT AND FUTURE STATE-OF-THE-ART REVIEW The Role of Nitroglycerin and Other Nitrogen Oxides in Cardiovascular Therapeutics Sanjay Divakaran, MD, Joseph Loscalzo, MD, PHD ABSTRACT The use of nitroglycerin in the treatment of angina pectoris began not long after its original synthesis in 1847. Since then, the discovery of nitric oxide as a biological effector and better understanding of its roles in vasodilation, cell permeability, platelet function, inammation, and other vascular processes have advanced our knowledge of the hemodynamic (mostly mediated through vasodilation of capacitance and conductance arteries) and nonhemodynamic effects of organic nitrate therapy, via both nitric oxidedependent and independent mechanisms. Nitrates are rapidly absorbed from mucous membranes, the gastrointestinal tract, and the skin; thus, nitroglycerin is available in a number of preparations for delivery via several routes: oral tablets, sublingual tablets, buccal tablets, sublingual spray, transdermal ointment, and transdermal patch, as well as intravenous formulations. Organic nitrates are commonly used in the treatment of cardiovascular disease, but clinical data limit their use mostly to the treatment of angina. They are also used in the treatment of subsets of patients with heart failure and pulmonary hypertension. One major limitation of the use of nitrates is the development of tolerance. Although several agents have been studied for use in the prevention of nitrate tolerance, none are currently recommended owing to a paucity of supportive clinical data. Only 1 method of preventing nitrate tolerance remains widely accepted: the use of a dosing strategy that provides an interval of no or low nitrate exposure during each 24-h period. Nitric oxides important role in several cardiovascular disease mechanisms continues to drive research toward nding novel ways to affect both endogenous and exogenous sources of this key molecular mediator. (J Am Coll Cardiol 2017;70:2393410) © 2017 by the American College of Cardiology Foundation. I n this review, we detail the discovery of nitro- glycerin and its early use in the treatment of angina; the history of the discovery of nitric oxide (NO), its sources, and its roles; the mechanism of action, preparations, and hemodynamic and non- hemodynamic effects of the organic nitrates, as well as their biotransformation; and the clinical uses and adverse effects of nitrate therapy, including toler- ance. We conclude by outlining current and future work investigating novel modulators of the NOsoluble guanylyl cyclase (sGC)cyclic guanosine monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio- vascular disease. EARLY HISTORY OF THE USE OF NITRATES IN CORONARY ARTERY DISEASE William Heberden is credited with coining the term angina pectorisin 1772 (1). Joseph Priestley, an English theologian and chemist, discovered NO 3 years later (2). Not until the next century, however, was NO and its congeners, organic nitrates, linked to the treatment of angina (3). Glyceryl trinitrate, or From the Department of Medicine, Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts. This work was supported in part by National Institutions of Health grants HL61795 and GM107618 (to Dr. Loscalzo). Both authors have re- ported that they have no relationships relevant to the contents of this paper to disclose. Manuscript received September 7, 2017; accepted September 19, 2017. Listen to this manuscripts audio summary by JACC Editor-in-Chief Dr. Valentin Fuster. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 70, NO. 19, 2017 ª 2017 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION PUBLISHED BY ELSEVIER ISSN 0735-1097/$36.00 https://doi.org/10.1016/j.jacc.2017.09.1064

Transcript of The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may...

Page 1: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

Listen to this manuscript’s

audio summary by

JACC Editor-in-Chief

Dr. Valentin Fuster.

J O U R N A L O F T H E AM E R I C A N C O L L E G E O F C A R D I O L O G Y V O L . 7 0 , N O . 1 9 , 2 0 1 7

ª 2 0 1 7 B Y T H E AM E R I C A N C O L L E G E O F C A R D I O L O G Y F O U N D A T I O N

P U B L I S H E D B Y E L S E V I E R

I S S N 0 7 3 5 - 1 0 9 7 / $ 3 6 . 0 0

h t t p s : / / d o i . o r g / 1 0 . 1 0 1 6 / j . j a c c . 2 0 1 7 . 0 9 . 1 0 6 4

THE PRESENT AND FUTURE

STATE-OF-THE-ART REVIEW

The Role of Nitroglycerin andOther Nitrogen Oxides inCardiovascular Therapeutics

Sanjay Divakaran, MD, Joseph Loscalzo, MD, PHD

ABSTRACT

Fro

wa

po

Ma

The use of nitroglycerin in the treatment of angina pectoris began not long after its original synthesis in 1847. Since then,

the discovery of nitric oxide as a biological effector and better understanding of its roles in vasodilation, cell permeability,

platelet function, inflammation, and other vascular processes have advanced our knowledge of the hemodynamic

(mostly mediated through vasodilation of capacitance and conductance arteries) and nonhemodynamic effects of organic

nitrate therapy, via both nitric oxide–dependent and –independent mechanisms. Nitrates are rapidly absorbed from

mucous membranes, the gastrointestinal tract, and the skin; thus, nitroglycerin is available in a number of preparations for

delivery via several routes: oral tablets, sublingual tablets, buccal tablets, sublingual spray, transdermal ointment,

and transdermal patch, as well as intravenous formulations. Organic nitrates are commonly used in the treatment of

cardiovascular disease, but clinical data limit their use mostly to the treatment of angina. They are also used in the

treatment of subsets of patients with heart failure and pulmonary hypertension. One major limitation of the use of nitrates

is the development of tolerance. Although several agents have been studied for use in the prevention of nitrate tolerance,

none are currently recommended owing to a paucity of supportive clinical data. Only 1 method of preventing nitrate

tolerance remains widely accepted: the use of a dosing strategy that provides an interval of no or low nitrate exposure

during each 24-h period. Nitric oxide’s important role in several cardiovascular disease mechanisms continues to drive

research toward finding novel ways to affect both endogenous and exogenous sources of this key molecular mediator.

(J Am Coll Cardiol 2017;70:2393–410) © 2017 by the American College of Cardiology Foundation.

I n this review, we detail the discovery of nitro-glycerin and its early use in the treatment ofangina; the history of the discovery of nitric

oxide (NO), its sources, and its roles; the mechanismof action, preparations, and hemodynamic and non-hemodynamic effects of the organic nitrates, as wellas their biotransformation; and the clinical uses andadverse effects of nitrate therapy, including toler-ance. We conclude by outlining current and futurework investigating novel modulators of the NO–

soluble guanylyl cyclase (sGC)–cyclic guanosinemonophosphate (cGMP) pathway that may result in

m the Department of Medicine, Brigham and Women’s Hospital, Harvar

s supported in part by National Institutions of Health grants HL61795 an

rted that they have no relationships relevant to the contents of this pape

nuscript received September 7, 2017; accepted September 19, 2017.

new therapeutic options in the treatment of cardio-vascular disease.

EARLY HISTORY OF THE USE OF NITRATES IN

CORONARY ARTERY DISEASE

William Heberden is credited with coining the term“angina pectoris” in 1772 (1). Joseph Priestley, anEnglish theologian and chemist, discovered NO 3years later (2). Not until the next century, however,was NO and its congeners, organic nitrates, linked tothe treatment of angina (3). Glyceryl trinitrate, or

d Medical School, Boston, Massachusetts. This work

d GM107618 (to Dr. Loscalzo). Both authors have re-

r to disclose.

Page 2: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

ABBR EV I A T I ON S

AND ACRONYMS

ALDH = aldehyde

dehydrogenase

BH4 = tetrahydro-L-biopterin

cAMP = cyclic adenosine

monophosphate

CAV1 = caveolin-1

cGMP = cyclic guanosine

monophosphate

EDRF = endothelium-derived

relaxing factor

eNOS = endothelial nitric oxide

synthase

iNOS = cytokine-inducible

nitric oxide synthase

nNOS = neuronal nitric oxide

synthase

NO = nitric oxide

NOS = nitric oxide synthase

P450 = cytochrome P450

enzyme(s)

sGC = soluble guanylyl cyclase

VEGF = vascular endothelial

growth factor

Divakaran and Loscalzo J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7

Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics N O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0

2394

nitroglycerin, was first synthesized byAscanio Sombrero in Turin, Italy. In 1847, henoted that “a very minute quantity put onthe tongue produced a violent headache forseveral hours” (4,5). Three year earlier, theFrench chemist Antoine Balard synthesizedamyl nitrite (4). Frederick Guthrie, an En-glish chemist, explored the actions of amylnitrite and published in 1859 that when itwas held near the nostrils, “after a lapse ofabout 50 seconds, a sudden throbbing of thearteries of the neck is felt, immediatelyfollowed by a flushing of neck, temples,and forehead and an acceleration action ofthe heart” (6).

T. Lauder Brunton, a Scottish physicianand medical scientist, first described theclinical effectiveness of amyl nitrite (4,5,7).Brunton began caring for patients withangina pectoris as a house physician at theEdinburgh Royal Infirmary (7). At the time,many treatments, including therapeuticbleeding, were being used to treat angina,largely unsuccessfully. In 1867, Bruntonpublished the first report of the use of amyl

nitrite in the treatment of angina pectoris (7,8). Hebelieved “the relief produced by [therapeutic]bleeding to be due to the diminution it occasioned inthe arterial tension” and “that a substance whichpossesses the power of lessening it in such aneminent degree as nitrite of amyl would probablyproduce the same effect, and might be repeatedas often as necessary without detriment to thepatient’s health” (7,8). In 1903, Charles-ÉmileFrançois-Franck, a French physiologist, firstsuggested that amyl nitrite was a coronary vasodi-lator (7).

In 1879, William Murrell described the symptom-atic effects of placing drops of 1% solution ofnitroglycerin in alcohol on the tongue (9). In addi-tion to reporting that it relieved angina and pre-vented subsequent attacks, he also reported thesymptoms he felt when he “[tried] its action on[himself]” (9). He described a “violent pulsation in[his] head” and noticed that his pulse was “muchfuller than natural” (9). In 1914, Brunton, whooriginally thought angina was caused by hyperten-sion, acknowledged that the “dilating action of amylnitrite and nitroglycerine upon the coronary vesselswould readily explain the relief they offer in anginapectoris, even in cases where the blood-pressure isnormal” (7).

NO IN THE CARDIOVASCULAR SYSTEM

HISTORY OF NO. In 1975, Diamond and Holmesshowed that tissue levels of cyclic adenosine mono-phosphate (cAMP) were increased in rat myometrialstrips maintained in a state of sustained contracture.They demonstrated that nitroglycerin could relax thedepolarized muscles without significantly increasingcAMP levels and, therefore, concluded that changesin total tissue levels of cAMP were not responsible forthe uterine relaxation caused by nitroglycerin (10).These investigators went on to show that nitroglyc-erin increased cyclic guanosine monophosphate(cGMP) levels in depolarized muscle (10). Thefollowing year, Diamond and Blisard showed that 200mmol/l nitroglycerin increased levels of cGMP by morethan 15-fold during relaxation of isolated strips ofphenylephrine-contracted canine femoral arterieswhile having no significant effect on cAMP levels (11).In 1977, pharmacologist Ferid Murad and his col-leagues published their seminal work on modulatingcontractility in bovine tracheal smooth muscleshowing that the guanylyl cyclase activators, sodiumnitrite, nitroglycerin, and sodium nitroprusside,increased cGMP levels and relaxed tracheal smoothmuscle (12). They went on to demonstrate that solu-tions of NO gas increased cGMP activity in soluble andparticulate preparations from various tissues in adose-dependent fashion, and that NO alone andin combination with sodium azide, sodium nitrite,hydroxylamine, and sodium nitroprusside increasedcGMP levels to approximately the same degree;they concluded that the 2 methods activateguanylyl cyclase through a “similar but undefinedmechanism” (13). Later, the same group postulatedthat “while the precise mechanism of guanylatecyclase activation by these agents is not known,activation may be due to the formation of NO oranother reactive material since NO also increasedguanylate cyclase activity” (14).

Working independently, Robert Furchgott andJohn Zawadzki published their observations on theimportance of the endothelium in blood vesselrelaxation in 1980 (4,15). They reported that acetyl-choline did not always produce blood vessel relaxa-tion in vitro, even though it was a potent vasodilatorin vivo. They found that loss of relaxation in vitrowas due to “unintentional rubbing of [the rabbitthoracic aorta’s] intimal surface against foreignsurfaces during its preparation.” When this mechan-ical injury was avoided during preparation of thetissue, it always relaxed in response to acetylcholine.

Page 3: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

FIGURE 1 Sources of NO

L-Arginine

Acidic environment(stomach, hypoxic/ischemic tissue)

Nitrate reductase(oral flora)

L-Citrulline NOS

NO

NO2–

NO3–

NO is a free radical that is synthesized by the family of NO synthases from L-arginine and

oxygen, yielding L-citrulline as a coproduct. In the blood vessel wall, NO is mainly

produced by endothelial nitric oxide synthase (NOS). There are 2 other isoforms of NOS

that also produce NO from L-arginine: neuronal NOS and cytokine-inducible NOS.

Additionally, bacterial flora present in the mammalian oral cavity is rich in nitrate

reductases and can convert dietary sources of nitrate to nitrite. In the extremely acidic

environment (pH #3) of the gastric lumen, protonation of nitrite can, in turn, yield

nitrous acid, which can spontaneously decompose to NO. This pathway of NO generation

is referred to as the enterosalivary nitrate circulation (nitrate-nitrite-NO pathway).

NO ¼ nitric oxide; NO2� ¼ inorganic nitrite; NO3

� ¼ inorganic nitrate.

J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7 Divakaran and LoscalzoN O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0 Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics

2395

Therefore, they concluded that acetylcholine-mediated relaxation of vascular smooth musclerequired the presence of endothelial cells (15).Furchgott and his colleagues eventually proposedthat bradykinin and other molecules acting on theendothelium caused relaxation of vascular smoothmuscle via a substance they termed endothelium-derived relaxing factor (EDRF) (16). The link to NO,however, was not yet appreciated (4).

Louis Ignarro and Salvador Moncada indepen-dently discovered that EDRF is NO in 1987 (4,17).After a series of experiments, Ignarro et al. (18,19)stated that “EDRF released from artery and veinpossesses identical biological and chemical propertiesas NO.” Moncada and colleagues suggested that EDRFand NO were “identical” as “NO released from endo-thelial cells is indistinguishable from EDRF in termsof biological activity, stability, and susceptibility toan inhibitor and to a potentiator” (20). Based on theircollective contributions to the field, the Nobel Prize inPhysiology or Medicine was awarded to Furchgott,Ignarro, and Murad in 1998 “for their discoveriesconcerning NO as a signaling molecule in the cardio-vascular system” (4).SOURCES OF NO. NO is a free radical that is synthe-sized by the family of NO synthases from L-arginineand oxygen, yielding L-citrulline as a coproduct(Figure 1) (21–23). In the blood vessel wall, NO ismainly produced by endothelial nitric oxide synthase(eNOS) (23,24). However, there are 2 other isoforms ofnitric oxide synthase (NOS) that also produce NO fromL-arginine: neuronal nitric oxide synthase (nNOS)and cytokine-inducible nitric oxide synthase (iNOS)(25–27). Although eNOS is the major NOS isoform thatregulates vascular function, both nNOS and iNOS areimplicated as sources in certain tissues and environ-ments (23). For example, vascular injury inducesexpression of nNOS in the neointima and medialsmooth muscle cells, and the production of iNOSwithin vascular smooth muscle cells after exposureto proinflammatory cytokines is a major cause ofvasodilation in sepsis (28,29).

S-nitrosothiols, such as S-nitrosoglutathione andS-nitrosohemoglobin, are also sources of NO (23,24).Under certain conditions, such as in the presenceof trace transition metal ions or photolysis,S-nitrosothiols decompose to liberate NO (30). Inaddition, S-nitrosothiols can undergo trans-S-nitrosation with other thiol groups and therebymodify cell or protein function (31–33).

Several proteins, such as hemoglobin, cytochromeP450 reductase, and cytochrome P450, can catalyze thereduction of nitrite or nitrate to generate NO (23).

Hemoglobin has enzymatic behavior as a nitritereductase under hypoxic conditions and is asensor and effector of hypoxic vasodilation (34). He-moglobin’s maximal nitrite reduction rate occurswhen hemoglobin is 40% to 60% saturated withoxygen (23,34). Cytochrome P450 reductase causes NOrelease by reducing nitrate, and can also facilitategeneration of S-nitrosothiols (35).

Bacterial flora present in the mammalian oralcavity is rich in nitrate reductases and can convertdietary sources of nitrate to nitrite. In the extremelyacidic environment (pH #3) of the gastric lumen,protonation of nitrite can, in turn, produce nitrousacid, which can spontaneously decompose to NO(36,37). This pathway of NO generation is referredto as the enterosalivary nitrate circulation (nitrate–nitrite–NO pathway) (Figure 1) and is a majorsource of NO (38,39).

NO’s ROLES. NO is a powerful vasodilator that in-duces formation of cGMP by activating sGC invascular smooth muscle cells (Figure 2) (13). cGMP canbind to and enhance protein kinase G activity, cGMP-gated ion channels, and cGMP-sensitive phosphodi-esterases (40). Protein kinase G promotes reuptake of

Page 4: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

FIGURE 2 Regulation of Vascular Tone by NO

L-Arginine

Endothelial Cell

Vascular SmoothMuscle Cell

Vascular Smooth MuscleRelaxation

Actin-MyosinATPase↓

Nitrovasodilators

Bioactivation

GTP

cGMPsGC

eNOS

cGMP-dep Kinase I

Ca2+

↓Ca2+

MLC-Pi

GMPPDE

MLCMLCK

MLCP

NO

NO

NO is a powerful vasodilator that induces formation of cGMP by activating soluble sGC in vascular smooth muscle cells. cGMP can bind to and enhance

protein kinase G activity, cGMP-gated ion channels, and cGMP-sensitive phosphodiesterases. Protein kinase G promotes reuptake of cytosolic calcium into

the sarcoplasmic reticulum, the movement of calcium from the intracellular to the extracellular environment, and the opening of calcium-activated

potassium channels. These changes result in reduction of vascular tone as the reduction in intracellular calcium impairs myosin light chain kinase’s ability

to phosphorylate myosin, resulting in smooth muscle cell relaxation. ATPase ¼ adenosine triphosphatase; Ca2þ ¼ calcium ion; cGMP ¼ cyclic guanosine

monophosphate; cGMP-dep Kinase I ¼ cyclic guanosine monophosphate-dependent protein kinase I; eNOS ¼ endothelial nitric oxide synthase;

GMP ¼ guanosine monophosphate; GTP ¼ guanosine triphosphate; MLC ¼ myosin light chain; MLCK ¼ myosin light chain kinase; MLCP ¼ myosin light

chain phosphatase; MLC-Pi ¼ phosphorylated myosin light chain; NO ¼ nitric oxide; PDE ¼ phosphodiesterase; sGC ¼ soluble guanylyl cyclase.

Divakaran and Loscalzo J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7

Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics N O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0

2396

cytosolic calcium into the sarcoplasmic reticulum, themovement of calcium from the intracellular to theextracellular environment, and the opening ofcalcium-activated potassium channels (23). Thesechanges result in reduction of vascular tone as thereduction in intracellular calcium impairs myosinlight chain kinase’s ability to phosphorylate myosin,resulting in smooth muscle cell relaxation (41).

NO has several other important roles in thevasculature. For example, when vascular endothelialgrowth factor (VEGF) binds to VEGF receptor-2, eNOSis activated and NO is formed (42–44). This NO isessential for VEGF function, as eNOS-deficient micehave markedly reduced vascular permeability andangiogenesis (45). S-nitrosylation of b-catenin by NOmodulates intercellular contacts between endothelialcells, likely accounting for NO-dependent changes inendothelial permeability (42). NO and organic nitratesalso impair platelet activation and aggregation by

activating platelet guanylyl cyclase and increasingintraplatelet cGMP (46–49).

Finally, a greater understanding of the conse-quences of impaired NO synthesis can yield moreinformation regarding the importance of NO’s role in awell-functioning cardiovascular system. For example,when low levels of its essential cofactor (6R)-5,6,7,8-tetrahydrobiopterin are present, eNOS producessuperoxide anion instead of NO in a process denotedenzymatic uncoupling (50). eNOS uncoupling has beenobserved in animal models of cardiovascular diseaseand in patients with cardiovascular risk factors, suchas hypertension and diabetes mellitus (51,52).

MECHANISM OF ACTION OF NITRATES

Organic nitrates, such as nitroglycerin, isosorbidedinitrate, and isosorbide mononitrate, are rapidlyabsorbed from several sites, such as the

Page 5: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

FIGURE 3 Chemical Structures of Key Nitrovasodilators

Isosorbidedinitrate

Isosorbidemononitrate Amyl nitriteNitroglycerin

Pentaerythrityltetranitrate

Sodiumnitroprusside Nicorandil

ONO2

ONO2

O2NO

O2NO

ONO2

ONO2

O2NO

ONO

O

O

ONO2

O2NO

H

H

ONO2

O

HN

N

O

O

OH

O2NO

H

H

2–

2Na+ CCN

N CC N

NN

O

N

C

Fe

Chemical structures of 7 key nitrovasodilators are shown.

J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7 Divakaran and LoscalzoN O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0 Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics

2397

gastrointestinal tract, mucous membranes, and skin,depending on the preparation (53). These compoundsare prodrugs with a nitrooxy (�O-NO2) moiety and aremetabolized to produce bioactive metabolites (54,55).The bioactivation of organic nitrates causes liberationof NO, allowing them to serve as NO donors (55,56).NO then causes vasodilation via its effect on vascularsmooth muscle cells, and impairs platelet activationas previously discussed (13).

Nitrates are also implicated in epigenetic regula-tion of vasodilation, including specifically smoothmuscle cell relaxation (57,58). Additionally, nitro-glycerin has been shown to increase the activity ofhistone acetylases with nitroglycerin-dependentvascular responses influenced by N 3-lysine acetyla-tion of contractile proteins (58).

PREPARATIONS

Nitrates are rapidly absorbed from mucous mem-branes, the gastrointestinal tract, and the skin (55).Thus, nitroglycerin is available in several prepara-tions for delivery via several routes: oral tablets,sublingual tablets, buccal tablets, sublingual spray,transdermal ointment, and transdermal patch; it isalso available in intravenous formulations, which are

used in hospitalized patients with angina, hyperten-sion, or heart failure (Figure 3, Table 1) (53,59).Nitroglycerin has a plasma half-life of approximately1 to 4 min; following hepatic and intravascularmetabolism, its biologically active metabolites have ahalf-life of approximately 40 min (53,60).

Isosorbide dinitrate is rapidly absorbed andundergoes extensive first-pass metabolism by theliver, which results in low bioavailability (53,55).Sustained-release formulations have a slower rate ofabsorption and can provide therapeutic plasmaconcentrations of the drug for up to 12 h (53).Oral isosorbide mononitrate is completely absorbedand has 100% bioavailability as it avoids first-passmetabolism. This leads to a more predictable doseresponse and plasma levels with less variation whencompared with other nitrates (55,61). Pentaerythrityltetranitrate is a long-acting organic nitrate availablein an oral formulation that acts within 10 to 20 minand has a duration of action of 8 to 12 h (57).Preclinical data suggested that pentaerythrityltetranitrate may not cause nitrate tolerance (see thefollowing text) or endothelial dysfunction; however,the drug is not currently commonly used, as clinicalstudies have not shown any clear benefit in patientswith chronic, stable angina (62).

Page 6: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

TABLE 1 Properties of Key Nitrovasodilators

Drug Route/Formulation DoseTime Until

Onset of ActionDuration of

Action

Nitroglycerin SL spray 0.4 mg 1–3 min 25 min

SL tablet 0.3–0.8 mg 1–3 min 25 min

TD patch 0.2–0.8 mg/h 30 min 10–12 h

IV infusion 5–400 mg/min Withinseconds

3–5 min

Isosorbide dinitrate PO IR tabletPO SR tablet

5–80 mg40–160 mg

60 min60 min

8 h12 h

Isosorbide mononitrate PO IR tabletPO SR tablet

5–20 mg30–240 mg

30–45 min30–45 min

6 h12–24 h

Amyl nitrite Inhalation ofampule

0.3 ml 30 s 3–15 min

Pentaerythrityltetranitrate

PO tablet 10–80 mg 10–20 min 8–12 h

Sodium nitroprusside IV infusion 0.3–10.0 mg/kg/min 1 min 6–12 min

Nicorandil PO tablet 5–20 mg 30–60 min 12 h

IR ¼ immediate release; IV ¼ intravenous; PO ¼ oral; SL ¼ sublingual; SR ¼ sustained release; TD ¼ transdermal.

Divakaran and Loscalzo J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7

Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics N O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0

2398

Nicorandil is a nicotinamide-nitrate ester whosechemical structure consists of a nicotinamide deriv-ative combined with a nitrate moiety (55). Nicorandilacts as both an NO donor and a Kþ

ATP channelopener to provide antianginal effects (63). The bio-activation of nicorandil occurs via the nicotinamide/nicotinic acid pathway and involves NO generationvia denitration (64). Nicorandil also dilates thecoronary microvasculature and peripheral resistancearterioles by causing vascular smooth cell hyperpo-larization and closure of L-type voltage-gatedcalcium channels via its action on Kþ

ATP channels;it is rapidly absorbed via the gastrointestinal tract,does not undergo first-pass metabolism, reachesmaximal plasma concentration after 30 to 60 min,and has a half-life of approximately 52 min(55,65,66). Nicorandil has an oral bioavailabilityof >75%, and its antianginal effects last approxi-mately 12 h (55,64,67).

Sodium nitroprusside, which is 44% cyanide byweight, comprises a ferrous ion center complexedwith 5 cyanic moieties and a nitrosyl group (68). It isavailable intravenously and interacts with oxyhemo-globin to produce methemoglobin and spontaneouslyrelease NO and cyanide (69,70). Sodium nitroprussidecauses direct venous and arterial vasodilation, is apotent pulmonary vasodilator, and is an inhibitor ofhypoxia-induced pulmonary vasoconstriction (68). Ithas an almost immediate onset of action and a veryshort half-life: its effects dissipate within 1 to 2 min(68).

Finally, there are several direct NO donors thatare already being used clinically, or have beensynthesized and studied for use particularly inbiomaterials. Inhaled NO gas diffuses rapidly across

the alveolar-capillary membrane and activates sGC inthe subjacent smooth muscle cells in the pulmonaryvasculature (71). N-diazeniumdiolates (NONOates) areNO donors that are synthesized by reacting primary orsecondary amines with NO gas at high pressure andlow temperatures; hydrolysis causes spontaneousdecomposition under physiological conditions andreleases NO (72–74). NO (as N2O3 or �ONOO) can reactwith thiols in vivo to form S-nitrosothiols, such asS-nitrosocysteine and S-nitrosoglutathione (75).S-nitrosothiol compounds can also be synthesizedchemically by reacting thiols with nitrous acid. Whenexposed to physiological fluids, S-nitrosothiolsdecompose to release NO (72). Other newer classes ofdirect NO donors that are actively being studiedinclude furoxans, benzofuroxans, and zeolites(76,77).

BIOTRANSFORMATION OF

ORGANIC NITRATES

Organic nitrates must undergo biotransformationto release a vasoactive molecule (78). The detailedstudy of the biotransformation of nitroglycerinhas suggested a high-potency pathway mediatedby aldehyde dehydrogenase (ALDH)-2 and a low-potency pathway mediated by other enzymes orlow-molecular-weight reductants (Figure 4) (78).

The high-potency pathway is important at clinicallyrelevant nitroglycerin concentrations (<1 mmol/l) andis localized to the mitochondria. The mitochondrialisoform of aldehyde dehydrogenase, ALDH-2, wasfound to be a key enzyme in the biotransformation ofnitroglycerin via this pathway as it generates inor-ganic nitrite and 1,2-glyceryl dinitrate from nitro-glycerin (79). There are 3 proposed mechanisms forthe release of the vasoactive molecule via thispathway: nitrogen oxides are formed via reduction ofinorganic nitrite; NO is formed directly in response tointeraction with ALDH-2; and inorganic nitritereleased from mitochondria may be reduced byxanthine oxidase in the cytoplasm to form NO (78).

The low-potency pathway is important at supra-pharmacological nitroglycerin concentrations (>1 mmol/l),is found in the smooth endoplasmic reticulum, andleads to formation of measurable amounts of NO invascular tissues (80,81). In this pathway, nitroglyc-erin is biotransformed by proteins such as deoxy-hemoglobin, deoxymyoglobin, cytochrome P450,xanthine oxidase, glutathione-S-transferase, glycer-aldehyde-3-phosphate dehydrogenase, or otherALDH isoforms; and by low-molecular-weight re-ductants such as cysteine, N-acetyl-cysteine, thio-salicylic acid, and ascorbate (57,82–84).

Page 7: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

FIGURE 4 Bioactivation of Organic Nitrates

Cyt Ox

XO

?

H+

ALDH2

low dose

high dose

sGC

cGMP cGK-I Relaxation

Mitochondrion

High potency nitratesGTN, PETN

PETriN

GDN, PEDN

NO2– NOx

.NO

.NO

.NO

P450 GMNPEMN

SmoothEndoplasmic Reticulum

Low potency nitratesISDN, ISDM

Organic nitrates must undergo biotransformation to release a vasoactive molecule via 1 of 2 pathways: a high-potency pathway mediated by ALDH-2, or a low-potency

pathway mediated by other enzymes or low-molecular-weight reductants. The high-potency pathway is important at clinically relevant nitroglycerin concentrations

(<1 mmol/l) and is localized to the mitochondria. The mitochondrial isoform of aldehyde dehydrogenase, ALDH-2, was found to be a key enzyme in the

biotransformation of nitroglycerin via this pathway as it generates inorganic nitrite (NO2�) and 1,2-glyceryl dinitrate from nitroglycerin. There are 3 proposed

mechanisms for the release of the vasoactive molecule via this pathway: nitrogen oxides are formed via reduction of inorganic nitrite; nitric oxide is formed directly in

response to interaction with ALDH-2; and inorganic nitrite released from mitochondria may be reduced by xanthine oxidase in the cytoplasm to form NO. The

low-potency pathway is important at suprapharmacological nitroglycerin concentrations (> 1 mmol/l), is found in the smooth endoplasmic reticulum, and leads to

formation of measurable amounts of NO in vascular tissues. In this pathway, nitroglycerin is biotransformed by proteins such as deoxyhemoglobin, deoxymyoglobin,

cytochrome P450, xanthine oxidase, glutathione-S-transferase, glyceraldehyde-3-phosphate dehydrogenase, or other ALDH isoforms; and by low-molecular-weight

reductants such as cysteine, N-acetyl-cysteine, thiosalicylic acid, and ascorbate. Reproduced with permission from Munzel and Daiber (78). ALDH2 ¼ aldehyde

dehydrogenase-2; cGK-I ¼ cyclic guanosine monophosphate-dependent protein kinase I; Cyt Ox ¼ cytochrome c oxidase; GDN ¼ 1,2-glyceryl dinitrate;

GMN ¼ 1,2-glyceryl mononitrate; GTN ¼ glyceryl trinitrate (nitroglycerin); Hþ ¼ hydrogen ion; ISDM ¼ isosorbide mononitrate; ISDN ¼ isosorbide dinitrate;

NO2- ¼ inorganic nitrite; NOx ¼ nitrogen oxides; P450 ¼ cytochrome P450 enzyme(s); PEDN ¼ pentaerythrityl dinitrate; PEMN ¼ pentaerythrityl mononitrate;

PETN ¼ pentaerythrityl tetranitrate; PETriN ¼ pentaerythrityl trinitrate; XO ¼ xanthine oxidase; other abbreviations as in Figure 2.

J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7 Divakaran and LoscalzoN O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0 Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics

2399

HEMODYNAMIC EFFECTS OF NITRATES

The organic nitrates have several hemodynamic ef-fects that are mostly mediated through vasodilationof capacitance veins and conductance arteries(Central Illustration) (53). In 1971, Parker et al. (85)published the results of a study of hemodynamicindices and coronary blood flow at rest and duringexercise before and after administration of nitro-glycerin in 15 patients with coronary artery diseaseand concluded that nitroglycerin acted primarily byreducing left ventricular oxygen requirementsthrough a reduction in left ventricular volume (85).NO-mediated dilation of capacitance veins decreasesventricular pre-load, which results in reduction inmyocardial oxygen demand. In 1975, Greenberg et al.(86), after angiographic and hemodynamic assess-ment of 10 patients, concluded that the mechanism ofaction of nitroglycerin “seems to relate best to thedecrease in systolic wall tension.” They also foundthat the end-diastolic wall tension decreased by 57%,

“suggesting the possibility that diastolic coronaryblood flow may be augmented by diminished extra-vascular resistance to flow” (86).

Nitrates also dilate large- and medium-sized coro-nary arteries and arterioles >100 mm in diameter. Thiseffect reduces left ventricular systolic wall tension viadecreasing afterload and, therefore, also decreasesmyocardial oxygen demand (53,87). The hemody-namic effects of nitrates on the coronary vasculature ofatherosclerotic patients also relieve angina. In 1972,Horwitz, Herman, and Gorlin published a study of 70patients (49 with coronary artery disease and 21without) with anginal chest pain and found that 76% ofpatients with coronary artery disease were regularlyrelieved of their pain within 3 min, while only 19%of those without coronary artery disease wereregularly relieved of their pain within 3 min (88).Nitrates dilate the epicardial coronary arteries,including stenotic segments, and also improve bloodflow in coronary collaterals via decreasing resistanceto collateral flow (87,89). In 1964, Fam and McGregor

Page 8: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

CENTRAL ILLUSTRATION Hemodynamic Effects of Nitroglycerin

Vasodilation of large- andmedium-sized coronaryarteries and arterioles

↓ Myocardial O2 Consumption

↓ Preload ↓ Left ventricularwall tension

↑ Subendocardialblood flow

Vasodilation ofcapacitance veins

Vasodilation of systemic andpulmonary arterial beds

Improvement in coronarycollateral blood flow

Dilation of stenoticepicardial coronary artery

segments

Vasodilation of conductancearteries

Epicardial coronary arterydilation

Nitroglycerin → NO

Divakaran, S. et al. J Am Coll Cardiol. 2017;70(19):2393–410.

The organic nitrates have several hemodynamic effects that are largely mediated through vasodilation of capacitance veins and conductance arteries. Seminal work in

the 1970s, as detailed in the main text, showed that nitroglycerin acts primarily by reducing left ventricular oxygen requirements through a reduction in left

ventricular volume. Nitric oxide (NO)–mediated dilation of capacitance veins decreases ventricular pre-load, which results in reduction in myocardial oxygen demand

and left ventricular wall tension. This results in an increase in subendocardial myocardial blood flow. Nitrates also dilate large- and medium-sized coronary arteries

and arterioles >100 mm in diameter. This effect reduces left ventricular systolic wall tension via decreasing afterload and, therefore, also decreases myocardial

oxygen demand. The hemodynamic effects of nitrates on the coronary vasculature also relieve angina. Nitrates dilate the epicardial coronary arteries, including

stenotic segments, and also improve blood flow in coronary collaterals via decreasing resistance to collateral flow. Nitrates, particularly sodium nitroprusside, can also

dilate the systemic arterial bed, and NO gas and sodium nitroprusside dilate the pulmonary vascular bed and inhibit hypoxia-induced pulmonary vasoconstriction.

O2 ¼ oxygen.

Divakaran and Loscalzo J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7

Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics N O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0

2400

studied the effects of nitroglycerin and dipyridamoleon coronary collateral blood flow in normal dogsand dogs with chronic myocardial ischemia producedby the placement of ameroid constrictors on the leftanterior descending and left circumflex arteries.Unlike dipyridamole, they found that nitroglycerin“did not cause a significant reduction in eitherretrograde flow or peripheral coronary pressure inspite of a greater fall in mean arterial pressure.”Additionally, when arterial pressure was heldconstant, nitroglycerin caused a large increase ofretrograde flow in the dogs with chronic myocardialischemia (90). Importantly, the organic nitrates do notdilate coronary microvessels that are <100 mm in

diameter, which reduces the risk of ischemia fromcoronary steal (91). Nitrates, particularly sodiumnitroprusside, can also dilate the systemic arterial bed,and NO gas and sodium nitroprusside dilate the pul-monary vascular bed and inhibit hypoxia-inducedpulmonary vasoconstriction (68).

NONHEMODYNAMIC EFFECTS OF NITRATES

The organic nitrates have also been found to haveseveral important, nonhemodynamic effects. Inhibi-tion of platelet function by organic nitrates was firstreported in 1967 by Hampton et al. (92), who showedthat nitroglycerin impairs platelet aggregation

Page 9: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7 Divakaran and LoscalzoN O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0 Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics

2401

in vitro. Subsequently, the organic nitrates have beenshown to inhibit platelet aggregation and functionby increasing intracellular cGMP and formingS-nitrosothiols, which are potent activators of sGCand inhibitors of platelet aggregation (46,93). Organicnitrates also have other antithrombotic effects, asactivation of sGC is accompanied by inhibition ofagonist-mediated calcium flux, which results in areduction of fibrinogen binding to the glycoproteinIlb/IIIa receptor of platelets (94).

Nitrates have also been found to have anti-inflammatory effects via NO’s role in the inflamma-tory process (95). NO inhibits neutrophil adhesionand chemotaxis in acute inflammation and modu-lates microvascular permeability (96–99). However,increased expression of iNOS has been implicated inchronic inflammatory conditions, as high-fluxNO fromiNOS or its derivates in the inflammatory milieu (e.g.,peroxynitrite) promotes adhesion molecule expres-sion and leukocyte–endothelial cell interactions,among other pro-inflammatory mechanisms (99).

Finally, nitroglycerin has been shown to induce aprotective phenotype that limits damage afterischemia and reperfusion. Nitroglycerin particularlyprotects against post-ischemic endothelial dysfunc-tion, in part by impairing the opening of the mito-chondrial permeability transition pore (100,101).

CLINICAL USES OF NITRATES

Organic nitrates, in intravenous, sublingual, and oralformulations, are often used in the management ofacute coronary syndrome (102,103). Treatment withnitrates causes vasodilation of the capacitance veinsand results in reduced ventricular filling pressure, walltension, and myocardial oxygen demand (53). As pre-viously discussed, nitrates also dilate the epicardialcoronary arteries, which improves coronary bloodflow, particularly in ischemic zones (87,89).

These well-demonstrated physiological effectsnotwithstanding, randomized controlled trial datasupporting a clinical outcome benefit for the use ofnitrates in acute coronary syndrome are lacking. TheGISSI-3 (Gruppo Italiano per lo Studio della Soprav-vivenza nell’Infarto Miocardico 3) trial randomlyassigned 19,394 patients with acute myocardialinfarction in a 2-by-2 factorial design to intravenousnitroglycerin followed by a nitrate patch or placebo,as well as to lisinopril or placebo. The primary studyendpoint of mortality at 6 weeks demonstrated nosignificant benefit of nitrate therapy. The subset ofpatients receiving combination therapy with lisino-pril and nitrates, however, had the lowest mortalityin the trial (104). At 6 months, there was no

difference in mortality in patients who received ni-trate therapy (105). The ISIS-4 (Fourth InternationalStudy of Infarct Survival) randomized 58,050 pa-tients presenting up to 24 h after the onset of asuspected acute myocardial infarction in a 2-by-2factorial design that involved treatment with capto-pril, isosorbide mononitrate, magnesium sulfate,and/or placebo. There was no significant reduction inmortality attributed to nitrate therapy (106). Of note,both the GISSI-3 and ISIS-4 trials were conducted inthe fibrinolytic era (i.e., prior to the percutaneouscoronary intervention era). By contrast, however, ameta-analysis of 10 trials, performed in the pre-fibrinolytic era (and pre-dating the GISSI-3 andISIS-4 trials), of patients randomized to intravenousnitroglycerin or sodium nitroprusside in acutemyocardial infarction showed a reduction in mortal-ity of 35% associated with nitrate therapy, with thegreatest reduction in mortality occurring during thefirst week of follow-up (107).

Nitroglycerin is the drug most frequently used totreat acute episodes of angina. It is usually given as asublingual tablet, but is also available as a sublingualspray. Sublingual nitroglycerin or nitroglycerin oralspray can also be used prior to angina-inducing ac-tivities to prevent the occurrence of acute angina(53,108,109).

Winsor and Berger (110) studied 53 patients withdocumented angina, and concluded in 1975 thatcontrolled-release oral nitroglycerin provided a sta-tistically significant clinical improvement of angina.In 1974, Reichek et al. (111) published their work on 14patients with angina pectoris and concluded thatnitroglycerin ointment produced a significant in-crease in exercise capacity, which persisted for atleast 3 h. Several studies have also shown animprovement in exercise capacity without angina viause of transdermal preparations (112–116). Takentogether, all of these studies support the use of oralorganic nitrates to increase exercise tolerance, pre-vent angina, and improve chronic stable angina. Incurrent practice, isosorbide dinitrate and isosorbidemononitrate are often used for these indications (53).

Although not available in the United States, nic-orandil is used in the treatment of chronic, stableangina in other countries. In the IONA (Impact ofNicorandil in Angina) trial, treatment with nicorandilin patients with stable angina statistically signifi-cantly reduced the primary endpoint of coronarydeath, nonfatal myocardial infarction, or unplannedhospitalization for angina (117).

The established treatment for Prinzmetal variantangina, or coronary artery spasm, is therapy withcalcium-channel blockers. Long-acting oral nitrates

Page 10: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

Divakaran and Loscalzo J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7

Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics N O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0

2402

have also been found to be helpful, and it is thoughtthat their vasodilatory effects are additive to calcium-channel blockade in this setting (118,119).

Intravenous nitroglycerin and sodium nitroprus-side are used to treat patients hospitalized with hy-pertensive urgency and emergency with preference tosodium nitroprusside, as it induces more arterialvasodilation than nitroglycerin. Although nitrateshave generally not been proven useful in the man-agement of hypertension, they are considered one ofthe drug classes-of-choice for patients with hyper-tension and stable angina (120). They should ideallybe used in combination with beta-blockers, or alone ifbeta-blockers are contraindicated or cause unaccept-able side effects (120).

Nitrates were previously considered first-line ther-apy for patients with acute heart failure with normal orhigh blood pressures owing to their previouslydescribed hemodynamic effects on the venous andarterial systems, pre-load, and afterload (121). A recentCochrane review, however, found no significant dif-ference between nitrate vasodilator therapy andalternative interventions for the treatment of acuteheart failure syndromes with regard to symptom reliefand hemodynamic variables (122). Given the strikingresults of the African-American Heart Failure Trialhowever, the combination of hydralazine and iso-sorbide dinitrate, together with angiotensin-converting enzyme inhibitors, beta-blockers, andaldosterone antagonists, is recommended by currentguidelines for African-Americans who require furtherblood pressure control and relief of symptoms fromNew York Heart Association functional class III or IVchronic heart failure (120,123,124). This combinationcan also be useful clinically in patients with chronicsystolic heart failure who cannot be given angiotensin-converting enzyme inhibitors or angiotensin receptorblockers (124).

Inhaled NO gas has several clinical uses in adults. Ithas a well-established role in vasoreactivity testing inpatients with pulmonary arterial hypertension. Thistesting is helpful to identify patients who mayrespond to therapy with calcium-channel blockers(71,125). Although robust data are lacking, inhaled NOgas is also used in patients both with and without aprior diagnosis of pulmonary hypertension with acutehypoxemic respiratory failure to improve ventilation-perfusion matching (126). Inhaled NO has an estab-lished role in the treatment of severe persistentpulmonary hypertension of the newborn, and arecently published Cochrane review also supports itsuse in the treatment of term and near-term infantswith hypoxic respiratory failure who do not have adiaphragmatic hernia (127,128). By contrast, a recent

Cochrane review does not support its use for respi-ratory failure in pre-term infants (129).

NITRATE TOLERANCE

One major problem with the use of nitrates is thedevelopment of tolerance, which is defined as “theloss of hemodynamic and anti-anginal effects duringsustained therapy” (53). Tolerance occurs followingchronic exposure to all nitrates and results in“complete or markedly diminished anti-anginal andanti-ischemic effects throughout the 24-h period oflong-term therapy” (130). In 1980, Thadani et al. (131)reported their studies of tolerance to oral isosorbidedinitrate and showed that both partial circulatorytolerance to isosorbide dinitrate and cross-toleranceto nitroglycerin developed rapidly during treatmentwith isosorbide dinitrate given every 6 h. They wenton to report similar findings regarding its antianginaleffect 2 years later (132). Subsequent studies reportedsimilar findings with different formulations of nitratetherapy (61,115,130,133–138).

The cause of nitrate tolerance is still incompletelyunderstood, but several hypotheses exist. One hy-pothesis argues that chronic treatment with organicnitrates triggers supersensitivity to vasoconstrictors,which attenuates the vasodilator effects of nitrates.This action is mediated by increased autocrine levelsof endothelin within the vasculature, with the sub-sequent activation of phospholipase C and proteinkinase C. These pathways lead to increased actomy-osin activity and myocyte contractility. Additionally,agonist-driven calcium-dependent activation of theRhoA/Rho kinase pathway contributes to vasocon-striction via inhibition of myosin light chain phos-phatase (139–141).

Another hypothesis for the mechanism ofnitrate tolerance is that chronic therapy with organicnitrates desensitizes sGC (142,143). S-nitrosylation ofsGC is a means by which “memory” of NO exposureis retained in smooth muscle cells, resulting indecreased responsiveness to NO, and could be amechanism of NO tolerance (139–144). Nitroglycerinmetabolism also promotes the production of reactiveoxygen species. Oxidation of thiol groups in theactive site of ALDH-2 by these reactive derivatives hasbeen observed during chronic nitroglycerin treat-ment. This post-translational modification may causeinhibition of ALDH-2 enzyme activity, which can leadto both reduced nitroglycerin biotransformation andefficacy (139,145–147).

Additionally, continuous treatment with nitro-glycerin has been shown to cause NO synthasedysfunction, likely through the reduced

Page 11: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7 Divakaran and LoscalzoN O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0 Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics

2403

bioavailability of tetrahydrobiopterin (148). NO syn-thase dysfunction can cause superoxide anion for-mation. If not effectively mitigated by superoxidedismutases, increased superoxide anion formationcan lead to the formation of peroxynitrite anion(through reaction with NO), which is a highly reactiveintermediate that promotes oxidant stress and re-duces NO bioavailability (139,149,150).

Nitrate therapy–induced increases in phosphodi-esterase activity have also been implicated in thedevelopment of tolerance (151). As previously dis-cussed, nitroglycerin is metabolized to NO, whichactivates sGC to increase cGMP. Phosphodiesterasedecreases cGMP levels, and, therefore, NO-inducedvasodilation is attenuated by its activity.

Finally, pseudotolerance also complicates chronictreatment with organic nitrates. This adaptive phe-nomenon is not considered true vascular tolerancebecause it occurs in response to every form of vaso-dilator therapy. Pseudotolerance is marked byneurohormonal activation, increased catecholaminerelease rates and circulating catecholamine levels,sodium retention, and intravascular volume expan-sion (139,152).

Although several agents have been studied for usein the prevention of nitrate tolerance, none arecurrently recommended due to a paucity of supportiveclinical data (130,136,153–160). Only 1 methodof preventing nitrate tolerance remains widelyaccepted: the use of a dosing strategy that provides aninterval of no or low nitrate exposure during each 24-hperiod (116,130,137,161–163). Nitrate tolerance israpidly reversed during a nitrate-free interval (53,164).

ADVERSE EFFECTS OF NITRATES

In addition to the development of tolerance, there areother important considerations to treatment withorganic nitrates. Common side effects of nitratetherapy include headache, flushing, lightheadedness,and postural hypotension. When sodium nitroprus-side is infused, it interacts with oxyhemoglobin toform methemoglobin and releases NO and cyanide.Therefore, patients receiving sodium nitroprussidemust be monitored for manifestations of both cyanidetoxicity, such as altered mental status, seizure, andmetabolic acidosis; and methemoglobinemia, suchas cyanosis, headache, fatigue, and lethargy(68,69,165,166). Cyanide toxicity from sodium nitro-prusside therapy is rare and is unlikely if the cumu-lative dose of nitroprusside does not exceed0.5 mg/kg/h. Large-infusion doses of nitroglycerincan also cause methemoglobinemia (167). There areseveral antidotes available to treat cyanide toxicity,

including sodium thiosulfate, sodium nitrite, amylnitrite, and hydroxocobalamin, as well as potentialantidotes that are undergoing development, such assulfanegen sodium. Sodium thiosulfate is availableintravenously, and acts as a sulfur donor to plasmarhodanase, an enzyme that transforms cyanide tothiocyanate, a nontoxic derivative (168). Intravenoussodium nitrite and inhaled amyl nitrite are methe-moglobin generators, and methemoglobin’s strongaffinity for cyanide binding is the rationale for theiruse as antidotes (169). Hydroxocobalamin is availableintravenously and contains a cobalt moiety, whichavidly binds to intracellular cyanide forming cyano-cobalamin (169–171). Sulfanegen sodium is a prodrugof 3-mercaptopyruvate that converts cyanide tothiocyanate (168,172). It was shown to be effective inreversing cyanide toxicity in a juvenile pig model, butwork is ongoing to develop it into an antidote that issafe for use in humans (173).

Additionally, there are certain clinical situationsin which nitrates should not be used, or should beused with extreme caution. Nitrates should notbe given to patients who have used a phosphodies-terase inhibitor, such as sildenafil or tadalafil, within24 to 48 h due to the risk of severe hypotension(174,175). Nitrates should also be avoided insuspected causes of right ventricular infarction, asnitrate-induced dilation of the venous capacitancebeds could cause hypotension given the need forhigh filling pressures in this setting (176). Patientswith hypertrophic cardiomyopathy should not beplaced on nitrate therapy, as it can increase outflowtract obstruction by decreasing pre-load andventricular volumes (177).

NOVEL MODULATORS OF THE

NO-sGC-cGMP PATHWAY

The organic nitrates have been and continue to bemainstays in the treatment of several acute andchronic cardiovascular conditions. Although many ofthe organic nitrates have been used clinically for de-cades, there are several new drugs and drug classesthat have been approved recently or are currentlybeing studied that have the potential to add to the listof modulators of the NO-sGC-cGMP pathway (178).Their methods of modulation of this pathway aresummarized in Figure 5.

Caveolin-1 (CAV1) is the main coat protein of cav-eolae, which are invaginations in the plasma mem-brane implicated in several biological processes,including signaling, in endothelial cells (178–182).eNOS activity is decreased when bound to CAV1(183,184). Cavnoxin, a peptide that activates eNOS,

Page 12: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

FIGURE 5 Novel Ways of Modulating the NOsGC-cGMP Pathway

IncreaseEndogenous

Cavnoxin

IntroduceCavnoxin-like

peptides

Enhancetranscription of

eNOS

Reverse eNOSuncoupling

Modulate theenterosalivary

nitrate circulation

Inhibit NADH-cytochrome b5

reductase 3

Disrupt hemoglobinα’s binding site for

eNOS

Fe2+-hemoglobin αto

Fe3+-hemoglobin α

Increase NO bioavailabilityIncrease endogenous NO production

eNOSactivation

Stimulate sGC

Increase cGMPproduction

Inhibitphosphodiesterases

Decrease cGMPbreakdown

Several novel modulators of the NO-sGC-cGMP pathway are undergoing active investigation, and the details are summarized in the main text. Methods of increasing

endogenous cavnoxin production or introducing synthesized cavnoxin-like peptides could increase endogenous NO production. Transcriptional enhances of eNOS, the

small molecules AVE3085 and AVE9488, have increased endogenous NO production, reversed eNOS uncoupling, and decreased eNOS production of superoxide anion in

mice. The enterosalivary nitrate circulation (nitrate-nitrite-NO pathway) is a source of NO that is derived from dietary inorganic nitrate intake, and production of NO

from this pathway is enhanced by hypoxia and acidosis. Research dedicated to exploring therapeutic options, such as prebiotics, probiotics, and antimicrobial agents,

that can modulate the microbiome and the nitrate-nitrite-NO pathway in heart failure, pulmonary hypertension, hypertension, obesity, and other cardiovascular

disease states is ongoing. Increasing the bioavailability of endogenous NO is another potential approach to modulating the NO-sGC-cGMP pathway. The oxidized form of

hemoglobin-a has a much lower affinity for NO than the reduced form, and, therefore, allows eNOS-generated NO to diffuse to underlying vascular smooth muscle

cells. Because NADH-cytochrome b5 reductase 3 reduces Fe3þ–hemoglobin-a to Fe2þ–hemoglobin-a, a potential way to increase NO bioavailability would be to inhibit

NADH-cytochrome b5 reductase 3. Another possible strategy would be to disrupt the binding site of hemoglobin-a for eNOS, and a small peptide, hemoglobin-aX, has

been developed as just such an inhibitor. Finally, modulating the NO-sGC-cGMP pathway in an NO-independent fashion is also being studied. The ciguats modulate sGC

activity to increase cGMP production independently of NO. The phosphodiesterases hydrolyze the phosphodiester bond of cGMP. Inhibition of these enzymes de-

creases cGMP breakdown. NADH ¼ nicotinamide adenine dinucleotide; Fe ¼ iron; other abbreviations as in Figure 2.

Divakaran and Loscalzo J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7

Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics N O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0

2404

was identified by studying the key residues in CAV1responsible for inhibition of eNOS function (185). Inwild-type mice, cavnoxin increases NO levels, causes areduction in vascular tone, and lowers systemic bloodpressure (185). Therefore, novel methods of increasingendogenous cavnoxin production or introducingsynthesized cavnoxin-like peptides could be a prom-ising way to increase endogenous NO production.

Another approach to increasing endogenous NO isto increase NO bioavailability by affecting hemoglo-bin-a’s ability to complex with eNOS. In the micro-circulation, hemoglobin-a forms a macromolecularcomplex with eNOS and controls the flux ofbioavailable NO (178,186). The reduced Fe2þ–O2–

hemoglobin-a reacts with NO rapidly and generatesnitrate and the oxidized Fe3þ–hemoglobin-a. This

oxidized form of hemoglobin-a has a much lower af-finity for NO than the reduced form, and, therefore,allows eNOS-generated NO to diffuse to underlyingvascular smooth muscle cells (178,187). Nicotinamideadenine dinucleotide (NADH)–cytochrome b5

reductase 3 reduces Fe3þ–hemoglobin-a to Fe2þ–hemoglobin-a. Therefore, yet another potentialway to increase NO bioavailability would beto inhibit NADH-cytochrome b5 reductase 3. Stillanother possible strategy would be to disrupt thebinding site of hemoglobin-a for eNOS; a small pep-tide, hemoglobin-aX, has been developed for thispurpose. Hemoglobin-aX has been found to decreaseblood pressure in mice and dilate constricted arteri-oles isolated from patients with hypertension(186,188).

Page 13: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7 Divakaran and LoscalzoN O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0 Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics

2405

Modulating the transcription, and therefore,translation of NOS is another focus of research anddevelopment efforts to augment levels of endoge-nous NO. Two small molecules, AVE3085 andAVE9488, have been identified as transcriptionalenhancers of eNOS that bind its promoter (178,189). Inapolipoprotein E knockout mice, 12 weeks of treat-ment with AVE9488 or AVE3085 reduced atheroscle-rotic plaque formation. Treatment of these mice withAVE9488 also reversed eNOS uncoupling, increasedvascular content of the essential eNOS cofactortetrahydro-L-biopterin (BH4), and reduced vascularcuff-induced neointima formation (189). Four weeksof treatment with AVE3085 was shown to attenuatecardiac remodeling in an experimental mouse modelinvolving aortic banding to induce remodeling (190).Finally, 9 weeks of treatment with AVE9488 wasshown to improve left ventricular remodeling andcontractile dysfunction in an experimental myocar-dial infarction rat model (191).

As referred to in the previous text with regard totreatment with AVE9488, reversing eNOS uncouplingis desired as it decreases eNOS production of super-oxide anion (50). Tetrahydrobiopterin has beenshown to improve endothelial dysfunction in patientswith type 2 diabetes mellitus and in patients oncyclosporine A after cardiac transplantation (52,192).However, a study of 49 patients randomized toreceive 400 mg/day of BH4, 700 mg/day of BH4, orplacebo for 2 to 6 weeks before coronary artery bypassgraft surgery found no effect of BH4 treatment onvascular function or superoxide production (193).More research is needed to explore alternative waysto reverse eNOS uncoupling and, thereby, increaseNO production and decrease superoxide anionproduction.

Ignarro et al. (194) showed that sGC activity couldbe modulated in an NO-independent fashion by pro-toporphyrin IX, an sGC activator. Protoporphyrin IXwas never used clinically due to its severe photo-sensitizing effect (178). However, a recently devel-oped class of drugs called the ciguats also modulatethe NO–sGC–cGMP pathway similarly in an NO-independent fashion (178). There are 2 subgroups ofciguats: NOsGC stimulators (which bind NOsGC andact through allosteric regulation) and NOsGC activa-tors (which occupy the heme binding site of NOsGCand work additively with NO) (178). Lificiguat was thefirst NOsGC stimulator that was identified when itwas found to stimulate sGC in rabbit platelets duringa small-molecule screen designed to identify novelplatelet aggregation inhibitors (195). Further work toimprove the solubility and efficacy of lificiguat led tothe development of riociguat, which is the only

NOsGC stimulator that is approved for clinicaluse (196). Riociguat is currently approved for thetreatment of pulmonary arterial hypertension andinoperable chronic thromboembolic pulmonary hy-pertension (178,197,198). There are ongoing trials ofriociguat’s potential role in the treatment of pulmo-nary hypertension and heart failure. Other NOsGC-stimulating ciguats that are being studied includevericiguat, which was shown to decreased circulatinglevels of N-terminal pro–B-type natriuretic peptidein patients with worsening chronic systolic heartfailure, as well as nelociguat, IW-1973, and IW-1701(178,199,200).

Cinaciguat was identified as an NOsGC activatorvia high-throughput screening (201,202). Its clinicaluse has been limited due to significant hypotension.In a placebo-controlled trial of 139 patients admittedwith acute, decompensated, systolic heart failure,cinaciguat significantly decreased pulmonary capil-lary wedge pressure, right atrial pressure, pulmo-nary vascular resistance, and systemic vascularresistance, and also significantly increased cardiacindex. However, the trial was stopped prematurelydue to an increased occurrence of hypotension atcinaciguat doses >200 mg/h (203). At lower doses,cinaciguat has been shown to decrease systemicblood pressure without improving dyspnea orcardiac index in patients with acute heart failure,therefore limiting its use in this population (204).Another NOsGC activator that is under developmentis ataciguat (178).

Another class of NOsGC-cGMP pathway modula-tors is the phosphodiesterases. These enzymesinhibit the pathway by hydrolyzing the phospho-diester bond of cGMP (178,205). There are 4 phos-phodiesterase 5 inhibitors in current clinical use thatinhibit cGMP breakdown: sildenafil, vardenafil,tadalafil, and avanafil. All 4 are approved for use inerectile dysfunction. Sildenafil and tadalafil are alsoused for pulmonary arterial hypertension, andtadalafil is approved for use in benign prostatichyperplasia (178). Work is ongoing to develop addi-tional phosphodiesterase inhibitors, including in-hibitors of nonselective phosphodiesterases andcGMP-selective phosphodiesterases (phosphodies-terases 5, 6, and 9) (178).

Finally, as discussed above, the enterosalivary ni-trate circulation (nitrate–nitrite–NO pathway) is asource of NO that is derived from dietary inorganicnitrate intake (38,39,206). Production of NO from thispathway is enhanced by hypoxia and acidosis(207,208). There is, therefore, interest in studying theuse of inorganic nitrates to improve exercise capacity,particularly in patients with heart failure with

Page 14: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

Divakaran and Loscalzo J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7

Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics N O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0

2406

preserved left ventricular ejection fraction (206). Inaddition, there is ongoing research dedicated toexploring therapeutic options, such as prebiotics,probiotics, and antimicrobial agents, that can modu-late the microbiome and the nitrate–nitrite–NOpathway in heart failure, pulmonary hypertension,hypertension, obesity, and other cardiovascular dis-ease states (39).

CONCLUSIONS

The use of nitrates in cardiovascular disease has along and storied history. They continue to play amajor role in current clinical practice to improve

symptoms despite the paucity of evidence of benefiton hard clinical endpoints. As research and develop-ment of new ways to modulate the NO-sGC-cGMPpathway continue, it may be time to revisit thestudy of nitrates in large, prospective clinical trials inthe percutaneous coronary intervention era.

ACKNOWLEDGMENT The authors thank Ms.Stephanie Tribuna for expert technical assistance.

ADDRESS FOR CORRESPONDENCE: Dr. Joseph Loscalzo,Department of Medicine, Brigham and Women’sHospital, 75 Francis Street, Boston, Massachusetts 02115.E-mail: [email protected].

RE F E RENCE S

1. Heberden W. Some account of a disorder of thebreast. Medical Transactions. London: Royal Col-lege of Physicians; 1772:59–67.

2. Priestley J. Experiments and observations ondifferent kinds of air. 2nd ed. London: J. Johnson,1775.

3. Steinhorn BS, Loscalzo J, Michel T. Nitroglycerinand nitric oxide—a rondo of themes in cardiovas-cular therapeutics. N Engl J Med 2015;373:277–80.

4. Marsh N, Marsh A. A short history of nitro-glycerine and nitric oxide in pharmacology andphysiology. Clin Exp Pharmacol Physiol 2000;27:313–9.

5. Berlin R. Historical aspects of nitrate therapy.Drugs 1987;33 Suppl 4:1–4.

6. Guthrie F. Contributions to the knowledge ofthe amyl group. 1. Nitryl of amyl and itsderivatives. J Chem Soc 1859;11:245–52.

7. Fye WB. T. Lauder Brunton and amyl nitrite: aVictorian vasodilator. Circulation 1986;74:222–9.

8. Brunton TL. On the use of nitrite of amyl inangina pectoris. Lancet 1867;2:97.

9. Murrell W. Nitro-glycerine as a remedy forangina pectoris. Lancet 1879;113:80–1.

10. Diamond J, Holmes TG. Effects of potassiumchloride and smooth muscle relaxants on tensionand cyclic nucleotide levels in rat myometrium.Can J Physiol Pharmacol 1975;53:1099–107.

11. Diamond J, Blisard KS. Effects of stimulant andrelaxant drugs on tension and cyclic nucleotidelevels in canine femoral artery. Mol Pharmacol1976;12:668–92.

12. Katsuki S, Murad F. Regulation of adenosinecyclic 3’,5’-monophosphate and guanosine cyclic3’,5’-monophosphate levels and contractility inbovine tracheal smooth muscle. Mol Pharmacol1977;13:330–41.

13. Arnold WP, Mittal CK, Katsuki S, Murad F. Ni-tric oxide activates guanylate cyclase and in-creases guanosine 3’:5’-cyclic monophosphatelevels in various tissue preparations. Proc NatlAcad Sci U S A 1977;74:3203–7.

14. Katsuki S, Arnold W, Mittal C, Murad F. Stim-ulation of guanylate cyclase by sodium nitroprus-side, nitroglycerin and nitric oxide in various tissuepreparations and comparison to the effects ofsodium azide and hydroxylamine. J Cyclic Nucle-otide Res 1977;3:23–35.

15. Furchgott RF, Zawadzki JV. The obligatory roleof endothelial cells in the relaxation of arterialsmooth muscle by acetylcholine. Nature 1980;288:373–6.

16. Cherry PD, Furchgott RF, Zawadzki JV,Jothianandan D. Role of endothelial cells inrelaxation of isolated arteries by bradykinin. ProcNatl Acad Sci U S A 1982;79:2106–10.

17. Ignarro LJ. Biological actions and properties ofendothelium-derived nitric oxide formed andreleased from artery and vein. Circ Res 1989;65:1–21.

18. Ignarro LJ, Buga GM, Wood KS, Byrns RE,Chaudhuri G. Endothelium-derived relaxing factorproduced and released from artery and vein is ni-tric oxide. Proc Natl Acad Sci U S A 1987;84:9265–9.

19. Ignarro LJ, Byrns RE, Buga GM, Wood KS.Endothelium-derived relaxing factor from pulmo-nary artery and vein possesses pharmacologic andchemical properties identical to those of nitricoxide radical. Circ Res 1987;61:866–79.

20. Palmer RM, Ferrige AG, Moncada S. Nitricoxide release accounts for the biological activity ofendothelium-derived relaxing factor. Nature 1987;327:524–6.

21. Loscalzo J. The identification of nitric oxide asendothelium-derived relaxing factor. Circ Res2013;113:100–3.

22. Stamler JS, Singel DJ, Loscalzo J. Biochemistryof nitric oxide and its redox-activated forms.Science 1992;258:1898–902.

23. Zhao Y, Vanhoutte PM, Leung SW. Vascularnitric oxide: Beyond eNOS. J Pharmacol Sci 2015;129:83–94.

24. Chen K, Pittman RN, Popel AS. Nitric oxide inthe vasculature: where does it come from andwhere does it go? A quantitative perspective.Antioxid Redox Signal 2008;10:1185–98.

25. Andrew PJ, Mayer B. Enzymatic function ofnitric oxide synthases. Cardiovasc Res 1999;43:521–31.

26. Alderton WK, Cooper CE, Knowles RG. Nitricoxide synthases: structure, function and inhibition.Biochem J 2001;357:593–615.

27. Förstermann U, Sessa WC. Nitric oxide syn-thases: regulation and function. Eur Heart J 2012;33:829–37. 837a–d.

28. Morishita T, Tsutsui M, Shimokawa H, et al.Vasculoprotective roles of neuronal nitric oxidesynthase. FASEB J 2002;16:1994–6.

29. Spink J, Cohen J, Evans TJ. The cytokineresponsive vascular smooth muscle cell enhancerof inducible nitric oxide synthase. Activation bynuclear factor-kappa B. J Biol Chem 1995;270:29541–7.

30. Singh RJ, Hogg N, Joseph J, Kalyanaraman B.Mechanismofnitric oxide release fromS-nitrosothiols.J Biol Chem 1996;271:18596–603.

31. Scharfstein JS, Keaney JF, Slivka A, et al.In vivo transfer of nitric oxide between a plasmaprotein-bound reservoir and low molecular weightthiols. J Clin Invest 1994;94:1432–9.

32. Liu Z, Rudd MA, Freedman JE, Loscalzo J.S-transnitrosation reactions are involved in themetabolic fate and biological actions of nitricoxide. J Pharmacol Exp Ther 1998;284:526–34.

33. Handy DE, Loscalzo J. Nitric oxide and post-translational modification of the vascular prote-ome: S-nitrosation of reactive thiols. ArteriosclerThromb Vasc Biol 2006;26:1207–14.

34. Huang Z, Shiva S, Kim-Shapiro DB, et al.Enzymatic function of hemoglobin as a nitritereductase that produces NO under allostericcontrol. J Clin Invest 2005;115:2099–107.

35. Li H, Liu X, Cui H, Chen YR, Cardounel AJ,Zweier JL. Characterization of the mechanism ofcytochrome P450 reductase-cytochrome P450-mediated nitric oxide and nitrosothiol generationfrom organic nitrates. J Biol Chem 2006;281:12546–54.

36. Maron BA, Tang SS, Loscalzo J. S-nitrosothiolsand the S-nitrosoproteome of the cardiovascularsystem. Antioxid Redox Signal 2013;18:270–87.

Page 15: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7 Divakaran and LoscalzoN O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0 Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics

2407

37. Spiegelhalder B, Eisenbrand G, Preussmann R.Influence of dietary nitrate on nitrite content ofhuman saliva: possible relevance to in vivo for-mation of N-nitroso compounds. Food CosmetToxicol 1976;14:545–8.

38. Lundberg JO, Weitzberg E, Gladwin MT. Thenitrate-nitrite-nitric oxide pathway in physiologyand therapeutics. Nat Rev Drug Discov 2008;7:156–67.

39. Koch CD, Gladwin MT, Freeman BA,Lundberg JO, Weitzberg E, Morris A. Enter-osalivary nitrate metabolism and the microbiome:intersection of microbial metabolism, nitric oxideand diet in cardiac and pulmonary vascular health.Free Radic Biol Med 2017;105:48–67.

40. Carvajal JA, Germain AM, Huidobro-Toro JP,Weiner CP. Molecular mechanism of cGMP-mediated smooth muscle relaxation. J Cell Phys-iol 2000;184:409–20.

41. Webb RC. Smooth muscle contraction andrelaxation. Adv Physiol Educ 2003;27:201–6.

42. Thibeault S, Rautureau Y, Oubaha M, et al.S-nitrosylation of beta-catenin by eNOS-derivedNO promotes VEGF-induced endothelial cellpermeability. Mol Cell 2010;39:468–76.

43. Papapetropoulos A, García-Cardeña G,Madri JA, Sessa WC. Nitric oxide production con-tributes to the angiogenic properties of vascularendothelial growth factor in human endothelialcells. J Clin Invest 1997;100:3131–9.

44. Ziche M, Morbidelli L, Choudhuri R, et al. Nitricoxide synthase lies downstream from vascularendothelial growth factor-induced but not basicfibroblast growth factor-induced angiogenesis.J Clin Invest 1997;99:2625–34.

45. Fukumura D, Gohongi T, Kadambi A, et al.Predominant role of endothelial nitric oxide syn-thase in vascular endothelial growth factor-induced angiogenesis and vascular permeability.Proc Natl Acad Sci U S A 2001;98:2604–9.

46. Loscalzo J. N-acetylcysteine potentiates inhi-bition of platelet aggregation by nitroglycerin.J Clin Invest 1985;76:703–8.

47. Mendelsohn ME, O’Neill S, George D,Loscalzo J. Inhibition of fibrinogen binding tohuman platelets by S-nitroso-N-acetylcysteine.J Biol Chem 1990;265:19028–34.

48. Pigazzi A, Heydrick S, Folli F, Benoit S,Michelson A, Loscalzo J. Nitric oxide inhibitsthrombin receptor-activating peptide-inducedphosphoinositide 3-kinase activity in humanplatelets. J Biol Chem 1999;274:14368–75.

49. Folts JD, Stamler J, Loscalzo J. Intravenousnitroglycerin infusion inhibits cyclic blood flowresponses caused by periodic platelet thrombusformation in stenosed canine coronary arteries.Circulation 1991;83:2122–7.

50. Förstermann U, Münzel T. Endothelial nitricoxide synthase in vascular disease: from marvel tomenace. Circulation 2006;113:1708–14.

51. Higashi Y, Sasaki S, Nakagawa K, et al. Tetra-hydrobiopterin enhances forearm vascularresponse to acetylcholine in both normotensiveand hypertensive individuals. Am J Hypertens2002;15:326–32.

52. Heitzer T, Krohn K, Albers S, Meinertz T.Tetrahydrobiopterin improves endothelium-dependent vasodilation by increasing nitric oxideactivity in patients with type II diabetes mellitus.Diabetologia 2000;43:1435–8.

53. Parker JD, Parker JO. Nitrate therapy for stableangina pectoris. N Engl J Med 1998;338:520–31.

54. Bogaert MG. Pharmacokinetics of organicnitrates in man: an overview. Eur Heart J 1988;9Suppl A:33–7.

55. Tarkin JM, Kaski JC. Vasodilator therapy:nitrates and nicorandil. Cardiovasc Drugs Ther2016;30:367–78.

56. Fung HL, Chung SJ, Bauer JA, Chong S,Kowaluk EA. Biochemical mechanism of organicnitrate action. Am J Cardiol 1992;70:4B–10B.

57. Daiber A, Münzel T. Organic nitrate therapy,nitrate tolerance, and nitrate-induced endothelialdysfunction: emphasis on redox biology andoxidative stress. Antioxid Redox Signal 2015;23:899–942.

58. Colussi C, Scopece A, Vitale S, et al. P300/CBPassociated factor regulates nitroglycerin-dependent arterial relaxation by N(ε)-lysine acet-ylation of contractile proteins. ArteriosclerThromb Vasc Biol 2012;32:2435–43.

59. den Uil CA, Brugts JJ. Impact of intravenousnitroglycerin in the management of acutedecompensated heart failure. Curr Heart Fail Rep2015;12:87–93.

60. Armstrong PW, Moffat JA, Marks GS. Arterial-venous nitroglycerin gradient during intravenousinfusion in man. Circulation 1982;66:1273–6.

61. Thadani U, Hamilton SF, Olson E, et al.Duration of effects and tolerance of slow-releaseisosorbide-5-mononitrate for angina pectoris. AmJ Cardiol 1987;59:756–62.

62. Münzel T, Meinertz T, Tebbe U, et al. Efficacyof the long-acting nitro vasodilator pentaerithrityltetranitrate in patients with chronic stable anginapectoris receiving anti-anginal background ther-apy with beta-blockers: a 12-week, randomized,double-blind, placebo-controlled trial. Eur Heart J2014;35:895–903.

63. Taira N. Nicorandil as a hybrid between ni-trates and potassium channel activators. Am JCardiol 1989;63:18J–24J.

64. Frydman A. Pharmacokinetic profile ofnicorandil in humans: an overview. J CardiovascPharmacol 1992;20 Suppl 3:S34–44.

65. Brodmann M, Lischnig U, Lueger A, Stark G,Pilger E. The effect of the Kþ agonist nicorandil onperipheral vascular resistance. Int J Cardiol 2006;111:49–52.

66. Akai K, Wang Y, Sato K, et al. Vasodilatoryeffect of nicorandil on coronary arterial micro-vessels: its dependency on vessel size and theinvolvement of the ATP-sensitive potassiumchannels. J Cardiovasc Pharmacol 1995;26:541–7.

67. Frydman AM, Chapelle P, Diekmann H, et al.Pharmacokinetics of nicorandil. Am J Cardiol 1989;63:25J–33J.

68. Friederich JA, Butterworth JF. Sodium nitro-prusside: twenty years and counting. Anesth Analg1995;81:152–62.

69. Smith RP, Kruszyna H. Nitroprusside producescyanide poisoning via reaction with hemoglobin.J Pharmacol Exp Ther 1974;191:557–63.

70. Ivankovich AD, Miletich DJ, Tinker JH. Sodiumnitroprusside: metabolism and general consider-ations. Int Anesthesiol Clin 1978;16:1–29.

71. Ichinose F, Roberts JD, Zapol WM. Inhaled ni-tric oxide: a selective pulmonary vasodilator: cur-rent uses and therapeutic potential. Circulation2004;109:3106–11.

72. Naghavi N, de Mel A, Alavijeh OS, Cousins BG,Seifalian AM. Nitric oxide donors for cardiovascu-lar implant applications. Small 2013;9:22–35.

73. Hrabie JA, Keefer LK. Chemistry of the nitricoxide-releasing diazeniumdiolate (“nitro-sohydroxylamine”) functional group and itsoxygen-substituted derivatives. Chem Rev 2002;102:1135–54.

74. Varu VN, Tsihlis ND, Kibbe MR. Basic sciencereview: nitric oxide—releasing prosthetic mate-rials. Vasc Endovascular Surg 2009;43:121–31.

75. Al-Sa’doni H, Ferro A. S-Nitrosothiols: a classof nitric oxide-donor drugs. Clin Sci (Lond) 2000;98:507–20.

76. Scatena R, Bottoni P, Pontoglio A, Giardina B.Pharmacological modulation of nitric oxiderelease: new pharmacological perspectives, po-tential benefits and risks. Curr Med Chem 2010;17:61–73.

77. Serafim RA, Primi MC, Trossini GH, Ferreira EI.Nitric oxide: state of the art in drug design. CurrMed Chem 2012;19:386–405.

78. Munzel T, Daiber A. Pharmacology of nitro-vasodilators. In: Bryan N, Loscalzo J, editors. Ni-trite and Nitrate in Human Health and Disease. 2ndedition. New York: Humana Press, 2017:349.

79. Chen Z, Zhang J, Stamler JS. Identification ofthe enzymatic mechanism of nitroglycerin bio-activation. Proc Natl Acad Sci U S A 2002;99:8306–11.

80. Kleschyov AL, Oelze M, Daiber A, et al. Doesnitric oxide mediate the vasodilator activity ofnitroglycerin? Circ Res 2003;93:e104–12.

81. Mülsch A, Bara A, Mordvintcev P, Vanin A,Busse R. Specificity of different organic nitrates toelicit NO formation in rabbit vascular tissues andorgans in vivo. Br J Pharmacol 1995;116:2743–9.

82. Münzel T, Daiber A, Mülsch A. Explaining thephenomenon of nitrate tolerance. Circ Res 2005;97:618–28.

83. Daiber A, Wenzel P, Oelze M, Münzel T. Newinsights into bioactivation of organic nitrates,nitrate tolerance and cross-tolerance. Clin ResCardiol 2008;97:12–20.

84. Daiber A, Münzel T, Gori T. Organic nitratesand nitrate tolerance—state of the art and futuredevelopments. Adv Pharmacol 2010;60:177–227.

85. Parker JO, West RO, Di Giorgi S. The effect ofnitroglycerin on coronary blood flow and the he-modynamic response to exercise in coronary arterydisease. Am J Cardiol 1971;27:59–65.

86. Greenberg H, Dwyer EM, Jameson AG,Pinkernell BH. Effects of nitroglycerin on themajor determinants of myocardial oxygen

Page 16: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

Divakaran and Loscalzo J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7

Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics N O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0

2408

consumption. An angiographic and hemodynamicassessment. Am J Cardiol 1975;36:426–32.

87. Brown BG, Bolson E, Petersen RB, Pierce CD,Dodge HT. The mechanisms of nitroglycerinaction: stenosis vasodilatation as a major compo-nent of the drug response. Circulation 1981;64:1089–97.

88. Horwitz LD, Herman MV, Gorlin R. Clinicalresponse to nitroglycerin as a diagnostic test forcoronary artery disease. Am J Cardiol 1972;29:149–53.

89. Goldstein RE, Stinson EB, Scherer JL,Seningen RP, Grehl TM, Epstein SE. Intraoperativecoronary collateral function in patients with cor-onary occlusive disease. Nitroglycerin responsive-ness and angiographic correlations. Circulation1974;49:298–308.

90. Fam M, McGregor M. Effect of coronaryvasodilator drugs on retrograde flow in areas ofchronic myocardial ischemia. Circ Res 1964;15:355–64.

91. Harrison DG, Bates JN. The nitrovasodilators.New ideas about old drugs. Circulation 1993;87:1461–7.

92. Hampton JR, Harrison MJ, Honour AJ,Mitchell JR. Platelet behaviour and drugs used incardiovascular disease. Cardiovasc Res 1967;1:101–7.

93. Schafer AI, Alexander RW, Handin RI. Inhibi-tion of platelet function by organic nitrate vaso-dilators. Blood 1980;55:649–54.

94. Loscalzo J. Antiplatelet and antithromboticeffects of organic nitrates. Am J Cardiol 1992;70:18B–22B.

95. Kumar S, Singh RK, Bhardwaj TR. Therapeuticrole of nitric oxide as emerging molecule. BiomedPharmacother 2017;85:182–201.

96. Lefer AM, Lefer DJ. Nitric oxide. II. Nitric oxideprotects in intestinal inflammation. Am J Physiol1999;276:G572–5.

97. Cherla RP, Ganju RK. Stromal cell-derivedfactor 1 alpha-induced chemotaxis in T cells ismediated by nitric oxide signaling pathways.J Immunol 2001;166:3067–74.

98. Adachi R, Matsui S, Kinoshita M, et al. Nitricoxide induces chemotaxis of neutrophil-like HL-60 cells and translocation of cofilin to plasmamembranes. Int J Immunopharmacol 2000;22:855–64.

99. Granger DN, Kubes P. Nitric oxide as antiin-flammatory agent. Methods Enzymol 1996;269:434–42.

100. Gori T, Di Stolfo G, Dragoni S, et al. Themechanism of nitrate-induced preconditioning.Clin Hemorheol Microcirc 2008;39:191–6.

101. Gori T, Di Stolfo G, Sicuro S, et al. Nitroglyc-erin protects the endothelium from ischaemia andreperfusion: human mechanistic insight. Br J ClinPharmacol 2007;64:145–50.

102. Karlberg KE, Saldeen T, Wallin R,Henriksson P, Nyquist O, Sylvén C. Intravenousnitroglycerin reduces ischaemia in unstable anginapectoris: a double-blind placebo-controlled study.J Intern Med 1998;243:25–31.

103. Curfman GD, Heinsimer JA, Lozner EC,Fung HL. Intravenous nitroglycerin in the treat-ment of spontaneous angina pectoris: a prospec-tive, randomized trial. Circulation 1983;67:276–82.

104. Gruppo Italiano per lo Studio della Soprav-vivenza nell’Infarto Miocardico. GISSI-3: effects oflisinopril and transdermal glyceryl trinitrate singlyand together on 6-week mortality and ventricularfunction after acute myocardial infarction. Lancet1994;343:1115–22.

105. Gruppo Italiano per lo Studio della Soprav-vivenza nell’Infarto Miocardico. Six-month effectsof early treatment with lisinopril and transdermalglyceryl trinitrate singly and together withdrawnsix weeks after acute myocardial infarction: theGISSI-3 trial. J Am Coll Cardiol 1996;27:337–44.

106. ISIS-4 (Fourth International Study of InfarctSurvival) Collaborative Group. ISIS-4: a rando-mised factorial trial assessing early oral captopril,oral mononitrate, and intravenous magnesiumsulphate in 58,050 patients with suspected acutemyocardial infarction. Lancet 1995;345:669–85.

107. Yusuf S, Collins R, MacMahon S, Peto R. Ef-fect of intravenous nitrates on mortality in acutemyocardial infarction: an overview of the rando-mised trials. Lancet 1988;1:1088–92.

108. Kimchi A, Lee G, Amsterdam E, Fujii K,Krieg P, Mason DT. Increased exercise toleranceafter nitroglycerin oral spray: a new and effectivetherapeutic modality in angina pectoris. Circula-tion 1983;67:124–7.

109. Sandler G, Ilahi MA, Lawson CW. Glyceryltrinitrate in angina pectoris. Lancet 1963;1:1130–6.

110. Winsor T, Berger HJ. Oral nitroglycerin as aprophylactic antianginal drug: clinical, physiologic,and statistical evidence of efficacy based on athree-phase experimental design. Am Heart J1975;90:611–26.

111. Reichek N, Goldstein RE, Redwood DR,Epstein SE. Sustained effects of nitroglycerinointment in patients with angina pectoris. Circu-lation 1974;50:348–52.

112. Scardi S, Camerini F, Pandullo C, Pollavini G.Efficacy of continuous and intermittent trans-dermal treatment with nitroglycerin in effortangina pectoris: a multicentric study. The Collab-orative Nitro Group. Int J Cardiol 1991;32:241–8.

113. Klemsdal TO, Gjesdal K. The effect of trans-dermal nitroglycerin on exercise tolerance in rela-tion to patch application time—a meta-analysis.Cardiovasc Drugs Ther 1992;6:641–9.

114. Parker JO, VanKoughnett KA, Fung HL.Transdermal isosorbide dinitrate in angina pecto-ris: effect of acute and sustained therapy. Am JCardiol 1984;54:8–13.

115. Thadani U, Hamilton SF, Olson E, et al.Transdermal nitroglycerin patches in angina pec-toris. Dose titration, duration of effect, and rapidtolerance. Ann Intern Med 1986;105:485–92.

116. DeMots H, Glasser SP. Intermittent trans-dermal nitroglycerin therapy in the treatment ofchronic stable angina. J Am Coll Cardiol 1989;13:786–95.

117. IONA Study Group. Effect of nicorandil oncoronary events in patients with stable angina: the

Impact Of Nicorandil in Angina (IONA) randomisedtrial. Lancet 2002;359:1269–75.

118. Stern S, Bayes de Luna A. Coronary arteryspasm: a 2009 update. Circulation 2009;119:2531–4.

119. Lombardi M, Morales MA, Michelassi C,Moscarelli E, Distante A, L’Abbate A. Efficacy ofisosorbide-5-mononitrate versus nifedipine inpreventing spontaneous and ergonovine-inducedmyocardial ischaemia. A double-blind, placebo-controlled study. Eur Heart J 1993;14:845–51.

120. Rosendorff C, Lackland DT, Allison M, et al.Treatment of hypertension in patients with coro-nary artery disease: a scientific statement from theAmerican Heart Association, American College ofCardiology, and American Society of Hypertension.J Am Coll Cardiol 2015;65:1998–2038.

121. Singh A, Laribi S, Teerlink JR, Mebazaa A.Agents with vasodilator properties in acute heartfailure. Eur Heart J 2017;38:317–25.

122. Wakai A, McCabe A, Kidney R, et al. Nitratesfor acute heart failure syndromes. CochraneDatabase Syst Rev 2013:CD005151.

123. Taylor AL, Ziesche S, Yancy C, et al. Combi-nation of isosorbide dinitrate and hydralazine inblacks with heart failure. N Engl J Med 2004;351:2049–57.

124. Yancy CW, Jessup M, Bozkurt B, et al. 2013ACCF/AHA guideline for the management of heartfailure: executive summary: a report of theAmerican College of Cardiology Foundation/American Heart Association Task Force on practiceguidelines. J Am Coll Cardiol 2013;62:e147–239.

125. Barst RJ, Gibbs JS, Ghofrani HA, et al. Upda-ted evidence-based treatment algorithm in pul-monary arterial hypertension. J Am Coll Cardiol2009;54:S78–84.

126. Hill NS, Preston IR, Roberts KE. Inhaledtherapies for pulmonary hypertension. Respir Care2015;60:794–802; discussion 802–5.

127. Barrington KJ, Finer N, Pennaforte T, Altit G.Nitric oxide for respiratory failure in infants bornat or near term. Cochrane Database Syst Rev 2017;1:CD000399.

128. Kinsella JP, Steinhorn RH, Krishnan US, et al.Recommendations for the use of inhaled nitricoxide therapy in premature newborns with severepulmonary hypertension. J Pediatr 2016;170:312–4.

129. Barrington KJ, Finer N, Pennaforte T. Inhalednitric oxide for respiratory failure in preterm in-fants. Cochrane Database Syst Rev 2017;1:CD000509.

130. Thadani U. Challenges with nitrate therapyand nitrate tolerance: prevalence, prevention, andclinical relevance. Am J Cardiovasc Drugs 2014;14:287–301.

131. Thadani U, Manyari D, Parker JO, Fung HL.Tolerance to the circulatory effects of oral iso-sorbide dinitrate. Rate of development and cross-tolerance to glyceryl trinitrate. Circulation 1980;61:526–35.

132. Thadani U, Fung HL, Darke AC, Parker JO. Oralisosorbide dinitrate in angina pectoris: comparisonof duration of action and dose-response relation

Page 17: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7 Divakaran and LoscalzoN O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0 Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics

2409

during acute and sustained therapy. Am J Cardiol1982;49:411–9.

133. Steering Committee, Transdermal Nitroglyc-erin Cooperative Study. Acute and chronic anti-anginal efficacy of continuous twenty-four-hourapplication of transdermal nitroglycerin. Am JCardiol 1991;68:1263–73.

134. Parker JO, Fung HL. Transdermal nitroglyc-erin in angina pectoris. Am J Cardiol 1984;54:471–6.

135. Thadani U, Lipicky RJ. Ointments and trans-dermal nitroglycerin patches for stable anginapectoris. Cardiovasc Drugs Ther 1994;8:625–33.

136. Thadani U, Lipicky RJ. Short and long-actingoral nitrates for stable angina pectoris. Car-diovasc Drugs Ther 1994;8:611–23.

137. Chrysant SG, Glasser SP, Bittar N, et al.Efficacy and safety of extended-release isosorbidemononitrate for stable effort angina pectoris. AmJ Cardiol 1993;72:1249–56.

138. Zimrin D, Reichek N, Bogin KT, et al. Anti-anginal effects of intravenous nitroglycerin over24 hours. Circulation 1988;77:1376–84.

139. Münzel T, Steven S, Daiber A. Organicnitrates: update on mechanisms underlying vaso-dilation, tolerance and endothelial dysfunction.Vascul Pharmacol 2014;63:105–13.

140. Münzel T, Giaid A, Kurz S, Stewart DJ,Harrison DG. Evidence for a role of endothelin 1and protein kinase C in nitroglycerin tolerance.Proc Natl Acad Sci U S A 1995;92:5244–8.

141. Oelze M, Knorr M, Kröller-Schön S, et al.Chronic therapy with isosorbide-5-mononitratecauses endothelial dysfunction, oxidative stress,and a marked increase in vascular endothelin-1expression. Eur Heart J 2013;34:3206–16.

142. Mülsch A, Busse R, Bassenge E. Desensitiza-tion of guanylate cyclase in nitrate tolerance doesnot impair endothelium-dependent responses. EurJ Pharmacol 1988;158:191–8.

143. Molina CR, Andresen JW, Rapoport RM,Waldman S, Murad F. Effect of in vivo nitroglycerintherapy on endothelium-dependent and indepen-dent vascular relaxation and cyclic GMP accumu-lation in rat aorta. J Cardiovasc Pharmacol 1987;10:371–8.

144. Sayed N, Baskaran P, Ma X, van den Akker F,Beuve A. Desensitization of soluble guanylylcyclase, the NO receptor, by S-nitrosylation. ProcNatl Acad Sci U S A 2007;104:12312–7.

145. Sydow K, Daiber A, Oelze M, et al. Centralrole of mitochondrial aldehyde dehydrogenaseand reactive oxygen species in nitroglycerintolerance and cross-tolerance. J Clin Invest 2004;113:482–9.

146. Daiber A, Oelze M, Coldewey M, et al.Oxidative stress and mitochondrial aldehydedehydrogenase activity: a comparison of pentaer-ythritol tetranitrate with other organic nitrates.Mol Pharmacol 2004;66:1372–82.

147. Wenzel P, Hink U, Oelze M, et al. Role ofreduced lipoic acid in the redox regulation ofmitochondrial aldehyde dehydrogenase (ALDH-2)activity. Implications for mitochondrial oxidativestress and nitrate tolerance. J Biol Chem 2007;282:792–9.

148. Gori T, Burstein JM, Ahmed S, et al. Folicacid prevents nitroglycerin-induced nitric oxidesynthase dysfunction and nitrate tolerance: a hu-man in vivo study. Circulation 2001;104:1119–23.

149. Hink U, Oelze M, Kolb P, et al. Role for per-oxynitrite in the inhibition of prostacyclin synthasein nitrate tolerance. J Am Coll Cardiol 2003;42:1826–34.

150. Warnholtz A, Mollnau H, Heitzer T, et al.Adverse effects of nitroglycerin treatment onendothelial function, vascular nitrotyrosine levelsand cGMP-dependent protein kinase activity inhyperlipidemic Watanabe rabbits. J Am Coll Car-diol 2002;40:1356–63.

151. Kim D, Rybalkin SD, Pi X, et al. Upregulationof phosphodiesterase 1A1 expression is associatedwith the development of nitrate tolerance. Circu-lation 2001;104:2338–43.

152. Parker JD, Farrell B, Fenton T, Cohanim M,Parker JO. Counter-regulatory responses tocontinuous and intermittent therapy with nitro-glycerin. Circulation 1991;84:2336–45.

153. Thadani U. Nitrate tolerance, rebound, andtheir clinical relevance in stable angina pectoris,unstable angina, and heart failure. CardiovascDrugs Ther 1997;10:735–42.

154. Katz RJ, Levy WS, Buff L, Wasserman AG.Prevention of nitrate tolerance with angiotensionconverting enzyme inhibitors. Circulation 1991;83:1271–7.

155. Boden WE, Finn AV, Patel D, Peacock WF,Thadani U, Zimmerman FH. Nitrates as an integralpart of optimal medical therapy and cardiacrehabilitation for stable angina: review of currentconcepts and therapeutics. Clin Cardiol 2012;35:263–71.

156. Dakak N, Makhoul N, Flugelman MY, et al.Failure of captopril to prevent nitrate tolerance incongestive heart failure secondary to coronaryartery disease. Am J Cardiol 1990;66:608–13.

157. Dupuis J, Lalonde G, Lemieux R, Rouleau JL.Tolerance to intravenous nitroglycerin in patientswith congestive heart failure: role of increasedintravascular volume, neurohumoral activationand lack of prevention with N-acetylcysteine. J AmColl Cardiol 1990;16:923–31.

158. Münzel T, Holtz J, Mülsch A, Stewart DJ,Bassenge E. Nitrate tolerance in epicardial arteriesor in the venous system is not reversed by N-acetylcysteine in vivo, but tolerance-independentinteractions exist. Circulation 1989;79:188–97.

159. Parker JO, Farrell B, Lahey KA, Rose BF.Nitrate tolerance: the lack of effect ofN-acetylcysteine. Circulation 1987;76:572–6.

160. Packer M, Lee WH, Kessler PD, Gottlieb SS,Medina N, Yushak M. Prevention and reversal ofnitrate tolerance in patients with congestive heartfailure. N Engl J Med 1987;317:799–804.

161. Fox KM, Dargie HJ, Deanfield J, Maseri A, forthe Transdermal Nitrate Investigators. Avoidanceof tolerance and lack of rebound with intermittentdose titrated transdermal glyceryl trinitrate. BrHeart J 1991;66:151–5.

162. Freedman SB, Daxini BV, Noyce D, Kelly DT.Intermittent transdermal nitrates do not improveischemia in patients taking beta-blockers or

calcium antagonists: potential role of reboundischemia during the nitrate-free period. J Am CollCardiol 1995;25:349–55.

163. Thadani U, Maranda CR, Amsterdam E, et al.Lack of pharmacologic tolerance and reboundangina pectoris during twice-daily therapy withisosorbide-5-mononitrate. Ann Intern Med 1994;120:353–9.

164. Parker JO, Fung HL, Ruggirello D, Stone JA.Tolerance to isosorbide dinitrate: rate of devel-opment and reversal. Circulation 1983;68:1074–80.

165. Cortazzo JA, Lichtman AD. Methemoglobi-nemia: a review and recommendations for man-agement. J Cardiothorac Vasc Anesth 2014;28:1043–7.

166. Coleman MD, Coleman NA. Drug-inducedmethaemoglobinaemia. Treatment issues. DrugSaf 1996;14:394–405.

167. Kaplan KJ, Taber M, Teagarden JR, Parker M,Davison R. Association of methemoglobinemia andintravenous nitroglycerin administration. Am JCardiol 1985;55:181–3.

168. Hottinger DG, Beebe DS, Kozhimannil T,Prielipp RC, Belani KG. Sodium nitroprusside in2014: A clinical concepts review. J AnaesthesiolClin Pharmacol 2014;30:462–71.

169. Cummings TF. The treatment of cyanidepoisoning. Occup Med (Lond) 2004;54:82–5.

170. Petrikovics I, Budai M, Kovacs K,Thompson DE. Past, present and future of cyanideantagonism research: From the early remedies tothe current therapies. World J Methodol 2015;5:88–100.

171. Thompson JP, Marrs TC. Hydroxocobalamin incyanide poisoning. Clin Toxicol (Phila) 2012;50:875–85.

172. Nagasawa HT, Goon DJ, Crankshaw DL,Vince R, Patterson SE. Novel, orally effectivecyanide antidotes. J Med Chem 2007;50:6462–4.

173. Belani KG, Singh H, Beebe DS, et al. Cyanidetoxicity in juvenile pigs and its reversal by a newprodrug, sulfanegen sodium. Anesth Analg 2012;114:956–61.

174. Kloner RA. Cardiovascular effects of the 3phosphodiesterase-5 inhibitors approved for thetreatment of erectile dysfunction. Circulation2004;110:3149–55.

175. Kostis JB, Jackson G, Rosen R, et al. Sexualdysfunction and cardiac risk (the Second PrincetonConsensus Conference). Am J Cardiol 2005;96:85M–93M.

176. Ondrus T, Kanovsky J, Novotny T, Andrsova I,Spinar J, Kala P. Right ventricular myocardialinfarction: from pathophysiology to prognosis.Exp Clin Cardiol 2013;18:27–30.

177. Zemanek D, Tomasov P, B�elehrad M, et al.Comparison of sublingual isosorbide dinitrate andValsalva maneuver for detection of obstruction inhypertrophic cardiomyopathy. Arch Med Sci 2015;11:751–5.

178. Kraehling JR, Sessa WC. Contemporary ap-proaches to modulating the nitric oxide-cGMPpathway in cardiovascular disease. Circ Res 2017;120:1174–82.

Page 18: The Role of Nitroglycerin and Other Nitrogen Oxides in ... · monophosphate (cGMP) pathway that may result in new therapeutic options in the treatment of cardio-vascular disease.

Divakaran and Loscalzo J A C C V O L . 7 0 , N O . 1 9 , 2 0 1 7

Nitroglycerin and Nitric Oxide in Cardiovascular Therapeutics N O V E M B E R 7 , 2 0 1 7 : 2 3 9 3 – 4 1 0

2410

179. Frank PG, Pavlides S, Lisanti MP. Caveolaeand transcytosis in endothelial cells: role inatherosclerosis. Cell Tissue Res 2009;335:41–7.

180. Gratton JP, Bernatchez P, Sessa WC. Cav-eolae and caveolins in the cardiovascular system.Circ Res 2004;94:1408–17.

181. Yu J, Bergaya S, Murata T, et al. Direct evi-dence for the role of caveolin-1 and caveolae inmechanotransduction and remodeling of bloodvessels. J Clin Invest 2006;116:1284–91.

182. Parton RG, Simons K. The multiple faces ofcaveolae. Nat Rev Mol Cell Biol 2007;8:185–94.

183. García-Cardeña G, Martasek P,Masters BS, et al. Dissecting the interactionbetween nitric oxide synthase (NOS) andcaveolin. Functional significance of the noscaveolin binding domain in vivo. J Biol Chem1997;272:25437–40.

184. Ju H, Zou R, Venema VJ, Venema RC. Directinteraction of endothelial nitric-oxide synthaseand caveolin-1 inhibits synthase activity. J BiolChem 1997;272:18522–5.

185. Bernatchez P, Sharma A, Bauer PM, Marin E,Sessa WC. A noninhibitory mutant of the caveolin-1 scaffolding domain enhances eNOS-derived NOsynthesis and vasodilation in mice. J Clin Invest2011;121:3747–55.

186. Keller TC, Butcher JT, Broseghini-Filho GB,et al. Modulating vascular hemodynamics with analpha globin mimetic peptide (HbaX). Hyperten-sion 2016;68:1494–503.

187. Straub AC, Lohman AW, Billaud M, et al.Endothelial cell expression of haemoglobin a

regulates nitric oxide signalling. Nature 2012;491:473–7.

188. Straub AC, Butcher JT, Billaud M, et al. He-moglobin a/eNOS coupling at myoendothelialjunctions is required for nitric oxide scavengingduring vasoconstriction. Arterioscler Thromb VascBiol 2014;34:2594–600.

189. Wohlfart P, Xu H, Endlich A, et al. Anti-atherosclerotic effects of small-molecular-weightcompounds enhancing endothelial nitric-oxidesynthase (eNOS) expression and preventing eNOSuncoupling. J Pharmacol Exp Ther 2008;325:370–9.

190. Chen Y, Chen C, Feng C, et al. AVE 3085, anovel endothelial nitric oxide synthase enhancer,attenuates cardiac remodeling in mice through theSmad signaling pathway. Arch Biochem Biophys2015;570:8–13.

191. Fraccarollo D, Widder JD, Galuppo P, et al.Improvement in left ventricular remodeling by theendothelial nitric oxide synthase enhancerAVE9488 after experimental myocardial infarc-tion. Circulation 2008;118:818–27.

192. Ramzy D, Rao V, Tumiati LC, et al. Tetrahy-drobiopterin prevents cyclosporine-induced vaso-motor dysfunction. Transplantation 2005;79:876–81.

193. Cunnington C, Van Assche T, Shirodaria C,et al. Systemic and vascular oxidation limits theefficacy of oral tetrahydrobiopterin treatment inpatients with coronary artery disease. Circulation2012;125:1356–66.

194. Ignarro LJ, Wood KS, Wolin MS. Activationof purified soluble guanylate cyclase by proto-porphyrin IX. Proc Natl Acad Sci U S A 1982;79:2870–3.

195. Ko FN, Wu CC, Kuo SC, Lee FY, Teng CM. YC-1, a novel activator of platelet guanylate cyclase.Blood 1994;84:4226–33.

196. Frey R, Mück W, Unger S, Artmeier-Brandt U,Weimann G, Wensing G. Single-dose pharmacoki-netics, pharmacodynamics, tolerability, and safetyof the soluble guanylate cyclase stimulator BAY63-2521: an ascending-dose study in healthy malevolunteers. J Clin Pharmacol 2008;48:926–34.

197. Ghofrani HA, Galiè N, Grimminger F, et al.Riociguat for the treatment of pulmonary arterialhypertension. N Engl J Med 2013;369:330–40.

198. Ghofrani HA, D’Armini AM, Grimminger F,et al. Riociguat for the treatment of chronicthromboembolic pulmonary hypertension. N EnglJ Med 2013;369:319–29.

199. Breitenstein S, Roessig L, Sandner P,Lewis KS. Novel sGC stimulators and sGC activa-tors for the treatment of heart failure. Handb ExpPharmacol 2017;243:225–47.

200. Gheorghiade M, Greene SJ, Butler J, et al.Effect of vericiguat, a soluble guanylate cyclasestimulator, on natriuretic peptide levels in patientswith worsening chronic heart failure and reduced

ejection fraction: the SOCRATES-REDUCED ran-domized trial. JAMA 2015;314:2251–62.

201. Stasch JP, Schmidt P, Alonso-Alija C, et al.NO- and haem-independent activation of solubleguanylyl cyclase: molecular basis and cardiovas-cular implications of a new pharmacological prin-ciple. Br J Pharmacol 2002;136:773–83.

202. Wunder F, Stasch JP, Hütter J, Alonso-Alija C, Hüser J, Lohrmann E. A cell-based cGMPassay useful for ultra-high-throughput screeningand identification of modulators of the nitricoxide/cGMP pathway. Anal Biochem 2005;339:104–12.

203. Erdmann E, Semigran MJ, Nieminen MS, et al.Cinaciguat, a soluble guanylate cyclase activator,unloads the heart but also causes hypotension inacute decompensated heart failure. Eur Heart J2013;34:57–67.

204. Gheorghiade M, Greene SJ, Filippatos G,et al. Cinaciguat, a soluble guanylate cyclaseactivator: results from the randomized, controlled,phase IIb COMPOSE programme in acute heartfailure syndromes. Eur J Heart Fail 2012;14:1056–66.

205. Uzunov P, Weiss B. Separation of multiplemolecular forms of cyclic adenosine-3’,5’-mono-phosphate phosphodiesterase in rat cerebellum bypolyacrylamide gel electrophoresis. Biochim Bio-phys Acta 1972;284:220–6.

206. Chirinos JA, Zamani P. The nitrate-nitrite-NOpathway and its implications for heart failure andpreserved ejection fraction. Curr Heart Fail Rep2016;13:47–59.

207. Maher AR, Milsom AB, Gunaruwan P, et al.Hypoxic modulation of exogenous nitrite-inducedvasodilation in humans. Circulation 2008;117:670–7.

208. Modin A, Björne H, Herulf M, Alving K,Weitzberg E, Lundberg JO. Nitrite-derived ni-tric oxide: a possible mediator of ’acidic-metabolic’ vasodilation. Acta Physiol Scand2001;171:9–16.

KEY WORDS angina, nitrate,nitrate-nitrite-NO pathway, nitric oxide,nitroglycerin, soluble guanylyl cyclase