nuevos medicamentos antihipertensivos

Upload: percovic

Post on 04-Apr-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    1/12

    http://cpt.sagepub.com/Therapeutics

    Journal of Cardiovascular Pharmacology and

    http://cpt.sagepub.com/content/15/3/257The online version of this article can be found at:

    DOI: 10.1177/1074248410368277

    2010 15: 257 originally published online 14 May 2010J CARDIOVASC PHARMACOL THERKatherine C. Wu and Gary Gerstenblith

    Review: Update on Newer Antihypertensive Medicines and Interventions

    Published by:

    http://www.sagepublications.com

    can be found at:Journal of Cardiovascular Pharmacology and TherapeuticsAdditional services and information for

    http://cpt.sagepub.com/cgi/alertsEmail Alerts:

    http://cpt.sagepub.com/subscriptionsSubscriptions:

    http://www.sagepub.com/journalsReprints.navReprints:

    http://www.sagepub.com/journalsPermissions.navPermissions:

    http://cpt.sagepub.com/content/15/3/257.refs.htmlCitations:

    What is This?

    - May 14, 2010OnlineFirst Version of Record

    - Sep 9, 2010Version of Record>>

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/content/15/3/257http://cpt.sagepub.com/content/15/3/257http://cpt.sagepub.com/content/15/3/257http://www.sagepublications.com/http://cpt.sagepub.com/cgi/alertshttp://cpt.sagepub.com/cgi/alertshttp://cpt.sagepub.com/subscriptionshttp://cpt.sagepub.com/subscriptionshttp://www.sagepub.com/journalsReprints.navhttp://www.sagepub.com/journalsReprints.navhttp://www.sagepub.com/journalsPermissions.navhttp://cpt.sagepub.com/content/15/3/257.refs.htmlhttp://online.sagepub.com/site/sphelp/vorhelp.xhtmlhttp://online.sagepub.com/site/sphelp/vorhelp.xhtmlhttp://online.sagepub.com/site/sphelp/vorhelp.xhtmlhttp://cpt.sagepub.com/content/early/2010/05/13/1074248410368277.full.pdfhttp://cpt.sagepub.com/content/early/2010/05/13/1074248410368277.full.pdfhttp://cpt.sagepub.com/content/15/3/257.full.pdfhttp://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://online.sagepub.com/site/sphelp/vorhelp.xhtmlhttp://cpt.sagepub.com/content/early/2010/05/13/1074248410368277.full.pdfhttp://cpt.sagepub.com/content/15/3/257.full.pdfhttp://cpt.sagepub.com/content/15/3/257.refs.htmlhttp://www.sagepub.com/journalsPermissions.navhttp://www.sagepub.com/journalsReprints.navhttp://cpt.sagepub.com/subscriptionshttp://cpt.sagepub.com/cgi/alertshttp://www.sagepublications.com/http://cpt.sagepub.com/content/15/3/257http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    2/12

    Review

    Update on Newer AntihypertensiveMedicines and Interventions

    Katherine C. Wu, MD,1 and Gary Gerstenblith, MD1

    Abstract

    The incidence and prevalence of systemic hypertension are reaching global epidemic proportions. Despite a diverse pharmacologicarmamentarium of agents to treat high blood pressure, suboptimal control remains a significant problem in as many as 43% ofpatients and this rate has not significantly improved over the past 2 decades. There are a variety of factors contributing to thisincluding patient nonadherence due to complex drug regimens and medication side effects, undertreatment, and treatment

    resistance. There, thus, remains a need to develop novel agents and approaches to antihypertensive therapy that facilitateattainment of optimal blood pressure levels. This monograph will review a number of new pharmacologic targets andinterventions as well as a novel method of drug delivery to patients.

    Keywords

    hypertension, antihypertensives, drug effects, reninangiotensin system

    It is estimated that as many as 1 in 3 adults in the United States

    have high blood pressure.1,2 Hypertension significantly

    increases the risk of myocardial infarction, stroke, heart failure,

    and end-stage renal disease. It thereby reduces overall life

    expectancy, accounting for close to 60 000 deaths per year1 and

    significant, lifelong disability for others. The direct and indirect

    costs of hypertension are expected to approach $74 billion in

    2009.1 Despite public health awareness and a multitude of

    existing treatments, achievement of optimal blood pressure

    control has minimally improved over the past 20 years and

    remains relatively low at 43%.3,4 In many patients with hyper-

    tension, adequate control may require 2 or more agents that tar-

    get various blood pressure regulatory systems, thereby more

    effectively and physiologically reducing blood pressure.5,6

    This results, in part, from the fact that the regulation of vascular

    tone and fluid balance is complex and involves many, often

    interrelated, pathways. The most important of these regulatory

    systems are the reninangiotensinaldosterone system

    (RAAS), the endothelin system, and the natriuretic peptides

    and kinin system. Although components of RAAS are targeted

    by such widely used antihypertensives as angiotensin-

    converting enzyme inhibitors (ACEIs) and angiotensin recep-

    tor blockers (ARBs), there remains a role for the development

    of therapeutic options with novel targets. In addition, as non-

    compliance contributes significantly to uncontrolled blood

    pressure, new drug delivery mechanisms, or agents that target

    more than one pathway in a single pill could be important ther-

    apeutically. This article will review newer drug classes and

    approaches to treating hypertension, several of which are still

    in development.

    Nebivolol

    Nebivolol is a third-generation b-adrenergic receptor blocker

    that has been available in Europe for a decade but was only

    Food and Drug Administration (FDA)approved for hyperten-

    sion treatment in the United States, in 2007.7-9 Compared to

    traditional b-blockers, it has a fundamentally different bio-chemical structure. Although there were potentially favorable

    effects in patients with angina and heart failure as well,7,10,11

    recently the FDA panel unanimously decided to deny approval

    to expand the drugs treatment indications to include chronic

    heart failure in the United States. Nebivolol is lipophilic and

    hence readily metabolized by the liver and able to reach all

    compartments of the body. It is very highly cardioselective,

    with 4 times higher affinity for b1 receptors as compared to

    metoprolol. It lacks both intrinsic sympathomimetic activity

    and membrane stabilizing activity. In addition, it does not

    appear to increase insulin resistance. Similar to carvedilol, it

    also has direct vasodilator properties. However, the mechan-isms of this differ and are instead mediated via stimulation of

    endothelial nitric oxide synthase activity, which results in

    increased nitric oxide bioavailability and decreased oxidative

    1 Division of Cardiology, Department of Medicine, Johns Hopkins University,

    Baltimore, MD, USA

    Corresponding Author:

    Katherine C. Wu, Division of Cardiology, Johns Hopkins Hospital, 600 N.

    Wolfe Street/Carnegie 568, Baltimore, MD 21287, USA

    Email: [email protected]

    Journal of Cardiovascular

    Pharmacology and Therapeutics

    15(3) 257-267

    The Author(s) 2010

    Reprints and permission:

    sagepub.com/journalsPermissions.nav

    DOI: 10.1177/1074248410368277

    http://cpt.sagepub.com

    257

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    3/12

    stress. Thus, nebivolol has the ability to improve endothelial

    dysfunction in addition to b-receptor blockade.

    The agent undergoes extensive first-pass hepatic metabo-

    lism by cytochrome P450 2D6 (CYP2D6), which yields multi-

    ple active metabolites. Although there is no effect clinically,

    how quickly the drug is metabolized is determined by genetic

    polymorphisms of 2D6. In extensive metabolizers, nebivolols

    half-life is 12 hours compared to 19 hours in slow metabolizers.

    Elimination is via both urinary and fecal routes. It is contrain-

    dicated in severe hepatic dysfunction and the starting dose

    requires reduction in patients with severe renal insufficiency

    or moderate liver impairment.

    Compared to placebo, 12-week therapy with nebivolol

    reduces diastolic blood pressure by an additional 5 to 8 mm

    Hg and systolic blood pressure by 2 to 7 mm Hg in patients

    with mild-to-moderate hypertension.12 In another trial of

    stages I-II hypertensive African American patients, nebivolol

    monotherapy significantly reduced diastolic and systolic pres-

    sures by an additional 2 to 7 mm Hg compared to placebo in a

    dose-dependent manner.13 Greater than 50% of the patients

    responded to treatment, which is noteworthy, given the data

    suggesting reduced effectiveness of traditional b-blockers in

    African Americans.

    Nebivolol has also been compared to otherb-blockers and

    antihypertensive drug classes. Its antihypertensive effects are

    similar to those of atenolol, bisoprolol, and metoprolol9,14,15

    Compared to ACEIs and ARBs, there is equivalent or some-

    what greater blood pressure lowering.16-18 Response rates

    are similar between nebivolol and calcium channel blockers.19

    Its adverse effects are similar to those of other traditional

    b-blockers and nebivolol is generally well tolerated. There are

    nonsignificant mild elevations of serum glucose levels, low-

    density lipoproteins (LDL), and triglycerides and nonsignifi-

    cant mild reductions in high-density lipoproteins (HDL).13,20

    Erectile function is reported to be unaffected and even poten-

    tially improved.9

    There are as yet, however, no large outcome trials of nebi-

    volol in hypertension, specifically in comparison to other

    agents. This is necessary, given the concern raised from

    meta-analyses regarding increased cardiovascular morbidity and

    mortality in hypertensive patients treated with b-blockade.7,21,22

    Hence, usingb-blockers as a class first-line agents to treat hyper-

    tension is generally not favored except in those circumstances

    wherein other properties of the b-blocker, for example, its

    anti-ischemic or secondary prevention benefits are desired. The

    future role that nebivolol will play in the decision making

    regarding which drugs to use in which hypertensive patients

    remains to be determined.

    Direct Renin Inhibitors

    The RAAS plays a critical role in the pathogenesis of hyperten-

    sion. Agents such as ACEIs, ARBs, and aldosterone receptor

    antagonists target various RAAS components (Figure 1) and

    are effective at reducing blood pressure and hypertensive com-

    plications. However, such agents act relatively downstream in

    the pathway and thus do not completely inhibit RAAS, because

    negative feedback loops lead to a compensatory rise in plasma

    renin concentration and subsequent elevations in plasma renin

    activity and angiotensin II levels, the so-called RAAS escape

    AngiotensinogenAngiotensinogen

    Angiotensin IAngiotensin-I

    Angiotensin IIAngiotensin-II

    Renin

    Angiotensin II

    receptors

    Angiotensin-II

    receptors

    ACE

    Renin inhibitors

    _

    ACEI_

    ARBs_

    KIDNEY

    Adrenal Gland

    Aldosterone release

    Aldosterone

    receptors

    Aldosterone

    receptors

    Aldosteronereceptorblockers

    _

    Angiotensin breakdown

    products

    Angiotensin breakdown

    products

    NEPinhibitors

    _

    ANP released

    from heart

    ANP released

    from heart

    NEPinhibitors

    Inhibit ANPbreakdown

    Figure 1. The reninangiotensin system and the sites of inhibition by renin-inhibitors, angiotensin-converting enzyme inhibitors (ACEIs),angiotensin receptor blockers (ARBs), aldosterone-receptor blockers, and neutral endopeptidase (NEP) inhibitors.

    258 Journal of Cardiovascular Pharmacology and Therapeutics 15(3)

    258

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    4/12

    phenomenon. Because the negative feedback suppression of

    angiotensin II on renin is disrupted by ACEI and ARB therapy,

    there is a compensatory rise in plasma renin and angiotensin

    I levels. Moreover, alternate, tissue-based, angiotensin-

    converting enzyme (ACE) -independent pathways are activated

    to convert angiotensin-I to angiotensin-II. Although the hemo-

    dynamic effects of RAAS escape result primarily from higherangiotensin-II levels, increased plasma renin may also contrib-

    ute via a direct vascular toxic effect.23 Hence, patients treated

    with ACEI and ARB may not achieve adequate blood pressure

    control with those agents alone. However, rigorous studies with

    robust clinical outcomes are ultimately required to adequately

    compare antihypertensive regimens to one another, despite

    theoretical advantages and concerns.24

    The incomplete blockade of RAAS with individual agents

    alone spurred further interest in combining therapies (dual-

    RAAS blockade) that inhibit different components of the

    RAAS pathway, thus potentially diminishing the effects of the

    escape phenomenon. Interrupting the RAAS cascade at its first-rate limiting step is potentially ideal because all downstream

    components would more effectively be suppressed. Renin con-

    trols the first and rate-limiting step of the RAAS and is secreted

    by the juxtaglomerular cells of the kidney. It cleaves specifi-

    cally and solely to angiotensinogen, which ultimately leads to

    angiotensin-II formation and subsequent angiotensin-II type 1

    receptor-mediated vasoconstriction and aldosterone release.

    Thus, inhibiting renin activity should block RAAS at its source

    of activation and thus provide more complete suppression of its

    downstream components, compared to other strategies.

    Development of oral renin inhibitors was limited for a num-

    ber of years due to its low bioavailability because of significanthepatic first-pass effects, as well as a short duration of action

    and low antihypertensive potency.25,26 In March 2007, aliski-

    ren was the first direct renin inhibitor to be approved by the

    US FDA for the treatment of essential hypertension as mono-

    therapy or in combination with other agents. It was designed

    using molecular-modeling techniques and crystal structure

    analysis.27,28 It is a nonpeptide, low-molecular-weight renin

    inhibitor which is still poorly absorbed (2.5% bioavailability)

    but is minimally metabolized and slowly excreted. It thus

    reaches a peak plasma concentration within 1 to 3 hours

    following oral administration. The accumulation half-life is

    *24 hours and steady state plasma levels are attained in 7 to

    8 days. The drug is primarily eliminated in the feces as unme-

    tabolized drug and

  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    5/12

    fibroblasts, and cardiomyocytes. Receptors for endothelin are

    widely distributed to varying degrees in many tissues and each

    cells response to endothelin varies, accounting for its multipli-

    city of action. Its vasoactive effects are mediated via interac-

    tion with 2 specific receptor subtypes, endothelin A (ETA

    )

    and endothelin B (ETB; Figure 2). The receptors have varying

    affinity for each of the 3 closely related isoforms of endothelin,

    ET-1, ET-2, and ET-3. Endothelin A has a >100-fold increased

    binding affinity for ET-1 than for ET-3. Endothelin-B has equal

    affinity for all 3 isoforms. Vascular tone is believed to be

    mediated via activation of ETA receptors found in vascular

    smooth muscle cells, which results in endothelins vaso-

    constrictive response. This effect is counterbalanced by ETBstimulation, which results in the release of vasodilatory

    endothelial-derived substances such as nitric oxide and prosta-

    cyclin. Endothelin receptors are also found abundantly in renal

    collecting duct cells and their stimulation regulates sodium

    reabsorption.41 Hence, the physiologic regulation of blood

    pressure is controlled, in part, by the equilibrium between ETAand ETB receptor stimulation and endothelins multiple path-

    ways affecting vascular tone and natriuresis.

    Much of what is known about the biologic effects of

    endothelin is derived from research with endothelin antago-

    nists. There has been a particularly large focus on the potential

    therapeutic benefits of such agents on pulmonary arterial

    hypertension, currently the only licensed clinical indication for

    endothelin receptor antagonists, which will not be reviewed

    here. However, several clinical trials also studied patients with

    essential hypertension.40 In 1998, bosentan, a mixed ETA and

    ETB receptor antagonist, was first compared with enalapril and

    placebo in the treatment of 293 patients with mild-to-moderate

    essential hypertension.42 Following 4 weeks of therapy, bosen-

    tan significantly reduced the primary end point, diatolic blood

    pressure, by 5.7 mm Hg, compared to the 1.8 mm Hg decline

    seen with placebo. The antihypertensive effect of bosentan was

    similar to that of enalapril therapy which resulted in a 5.8 mm

    Hg decline in diastolic blood pressure. This occurred without

    evidence of reflex tachycardia or of sympathetic nervous sys-

    tem or RAAS activation, which can characterize the response

    to vasodilators. Although relatively well tolerated and safe,

    *7% of the bosentan-treated patients experienced asympto-

    matic elevations of liver transaminases to greater than twice the

    upper limit of normal, which normalized after discontinuing

    the therapy. This effect appeared somewhat dose dependent.

    Several recent clinical trials have examined darusentan, a

    selective ETA receptor antagonist, which would be expected

    to be pathophysiologically superior to combined ETA/ETBreceptor antagonist because the vasodilatory effects of ETBreceptor stimulation would not be suppressed. Phase II studies

    in moderate and resistant hypertension were promising with a

    dose-dependent decrease in blood pressure that was greater

    than that achieved by bosentan.43 An initial phase III trial was

    recently published in September 2009 and involved 379

    patients with treatment-resistant hypertension already receiv-

    ing at least 3 other antihypertensive agents.44 The primary end

    point was a change in seated systolic and diastolic blood pres-

    sure over 14 weeks of therapy. Darusentan significantly

    reduced the systolic and diastolic blood pressures by an addi-

    tional 10 mm Hg compared to placebo. This antihypertensive

    effect was accompanied by a significantly increased fluid

    ENDOTHELIALCELLENDOTHELIAL CELL

    Endothelin-1

    ECEETB

    VASCULAR SMOOTH MUSCLE CELL

    ETB

    Big Endothelin-1

    Increased NO

    Vasoconstriction

    ETA

    KIDNEY

    Vasoconstriction

    Salt and water

    retention

    Vasodilation

    Figure 2. The opposing effects of endothelin on endothelial and vascular smooth muscle cells are shown and are mediated by differentialresponse to endothelin-receptor (ETA and ETB) stimulations. Endothelin-converting enzyme (ECE) is found on endothelial cells and isresponsible for conversion of big endothelin-1 to endothelin-1, the predominant form of endothelin.

    260 Journal of Cardiovascular Pharmacology and Therapeutics 15(3)

    260

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    6/12

    retention, most likely related to the vasodilatory effects of

    darusentan and that required higher doses of diuretics.

    However, these results have since been supplanted by a

    larger phase III trial in 849 resistant hypertension patients,

    which failed to demonstrate a significant change in trough sys-

    tolic and diastolic blood pressure at 14 weeks, the prespecified

    primary endpoints, compared with placebo. Other secondaryend points were reportedly met and included changes in ambu-

    latory blood pressure, percentage of patients reaching systolic

    blood pressure goal, and change in estimated glomerular filtra-

    tion rate. Nonetheless, because of the negative primary end

    point results, the drugs sponsor anticipates that it will no lon-

    ger develop the agent45 and it is currently unclear what role, if

    any, endothelin-receptor antagonists will play in future studies

    of systemic hypertension.

    Vasopeptidase Inhibition

    Neutral endopeptidase or neprylisin (NEP) is a zinc-dependentmetallopeptidase found on endothelial cells, vascular smooth

    muscle cells, cardiac myocytes, renal epithelial cells, and fibro-

    blasts.46 It is responsible for the degradation of natriuretic pep-

    tides such as atrial natriuretic peptide (ANP; Figure 1), brain

    natriuretic peptide (BNP), and C-type natriuretic peptide

    (CNP). Atrial natriuretic peptide and BNP are released from the

    myocardium in response to myocardial stretch, CNP is found in

    the kidney, heart, lung, and vascular endothelium and released

    in response to shear stress. Natriuretic peptides promote

    natriuresis, diuresis, and vasorelaxation and are antiprolifera-

    tive. However, NEP is also required for the metabolism of

    angiotensin-II (Figure 1). Hence, NEP inhibitors used as mono-therapy induce natriuresis and potentially reduce blood pres-

    sure because they increase circulating ANP levels. However,

    this antihypertensive effect is offset variably by increased

    angiotensin-II levels, which may explain the lack of net antihy-

    pertensive effect seen in small human trials of NEP inhibitors

    alone.47,48 This subsequently led to interest in developing

    dual-acting compounds that could block both NEP and the gen-

    eration or activity of angiotensin II.

    The other 2 main enzyme regulators in RAAS are ACE and

    endothelin-converting enzyme (ECE; Figure 2), both of which

    are also zinc metallopeptidases. Neutral endopeptidase shares

    similarities in structure and mechanism of action with ACE and

    ECE. Hence, it has been possible to design molecules that

    simultaneously target both ACE/NEP and NEP/ECE (dual

    vasopeptidase inhibitors) as well as ACE/NEP/ECE (triple

    vasopeptidase inhibitors).49 Such a multipronged approach

    with a single molecule could theoretically improve antihyper-

    tensive efficacy and compliance without the need for multiple

    agents and complex dosing regimens.

    Dual vasopeptidase inhibition has had limited success. By

    inhibiting both ACE and NEP, there are synergistic effects with

    greater blood pressure lowering not seen with selective inhibi-

    tors alone.49,50 There is increased activity of the endogenous

    vasodilator systems and decreased angiotensin-II production,

    which result in greater hemodynamic and renal effects.

    However, clinical trials of dual inhibition using the agent

    omapatrilat were associated with an increased risk of angioe-

    dema, compared to ACE-inhibition alone, approaching 2% to

    6%.49-52 The mechanism for this is not completely delineated but

    has caused the cessation of further development of omapatrilat.

    Another dual ACE/NEP inhibitor, ilepatril (AVE-7688), has

    undergone phase II testing in patients with mild-to-moderatehypertension and was found to lower blood pressure more effec-

    tively than losartan and had a reduced, 1.07%, incidence of

    angioedema.53 Phase IIb/III clinical trials are underway.

    The increased rate of angioedema associated with dual

    ACE/NEP inhibition is postulated to be due to inhibition of

    aminopeptidase P (APP) which along with ACE inhibits the

    breakdown of substance P and bradykinin, with possibly only

    a minor contribution specifically from NEP inhibition. Thus,

    a combined ARB and NEP inhibitor is postulated to be safer

    with a lower probability of angioedema because APP and ACE

    levels would not be affected. The pharmacokinetics of such an

    agent were recently tested in preclinical studies and in healthyvolunteers.54 It was found to be well tolerated in humans with

    no serious adverse effects, but additional longer term clinical

    studies are required to determine the incidence of angioedema

    and its efficacy in patients with hypertension. Dual NEP/ECE

    inhibition has also been studied, predominantly in heart failure.

    However, in this patient population, such inhibitors failed to

    reduce systemic blood pressure,55 and hence either triple

    ACE/NEP/ECE inhibition or concomitant therapy with ACEI

    or ARBs is needed.

    Triple ACE/NEP/ECE inhibitors are still in preclinical devel-

    opment and their toxicity profiles are currently unknown.46,49

    The advantage of such agents is in the potential for reducingpolypharmacy. However, the broad targeting of such drugs may

    predispose to adverse effects because many other downstream

    enzymatic pathways could be affected. Careful, objective eva-

    luation of this pharmacologic class is needed but may be war-

    ranted, given their theoretical benefits.

    Antihypertensive Effects of Novel Progestins

    The Womens Health Initiative reported an increased risk of

    cardiovascular disease in those receiving postmenopausal

    estrogen and progesterone replacement therapy (hormone

    replacemant therapy [HRT]). This resulted in changes in the

    guidelines for prescribing HRT and decreased use.56 However,

    a significant number of women experience major unacceptable

    symptoms associated with estrogen deficiency accompanying

    menopause, in addition to the increased cardiovascular risk

    of the postmenopausal state itself. Hence, alternative strategies

    for hormone therapy have been developed. A novel progestin,

    drospirenone, was synthesized from 17-a spironolactone, an

    aldosterone-receptor blocker (Figure 1) which has a high-

    affinity for aldosterone receptors57,58 and which is used for the

    treatment of hypertension. It has both antialdosterone and anti-

    androgenic effects and could thus reduce salt and water reten-

    tion and hence cause less weight gain and hypertension, as

    compared to traditional HRT. Drosperinone is rapidly and

    Wu and Gerstenblith 261

    261

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    7/12

    completely absorbed and metabolized following oral ingestion.

    Metabolism is independent of the cytochrome P450 system.

    Peak serum concentration is achieved within 1 hour of admin-

    istration and steady state is achieved in 10 days. Elimination

    half-life is 30 hours and excretion of the drugs metabolites are

    via the feces and urine. Contraindications include severe renal

    insufficiency or acute renal failure.Drosperinone has been studied in postmenopausal women

    with stages I-II hypertension in 2 clinical trials of relatively

    short duration (8 and 12 weeks).59,60 There were statistically

    significant reductions, on the order of 3 mm Hg to 5 mm Hg,

    in both 24-hour ambulatory and clinic blood pressures com-

    pared to placebo. The drug was generally well tolerated with

    an adverse event rate of 6.9%. Dizziness was the most common

    adverse event at 4% compared to 2% in placebo. There was no

    clinically significant increase in serum potassium values.

    Drosperinone currently is approved for moderate to severely

    abnormal vasomotor function and atrophic vulva. It is unlikely

    to be effective as a single antihypertensive agent, given itsrather modest blood pressure-lowering ability. However, its

    mechanism of action may support its use in postmenopausal

    women with hypertension, though more extensive efficacy and

    studies comparing it with other HRT options are needed.57,61

    Hypertension Vaccines

    A significant deterrent to optimal blood pressure control is

    patient noncompliance with medications, the rate of which may

    approach as high as 55% at 3 years following initiation of an

    antihypertensive therapy.62 Hence, the concept of a vaccine

    that eliminates the need to take medications is an attractiveoption and has been explored since the 1950s.63 Vaccine efforts

    have targeted components of RAAS. Initial preclinical efforts

    were either ineffective in reducing blood pressure or required

    an immunologic adjuvant that is toxic to and prohibited for use

    in humans.

    Recent attempts at vaccine development are directed at

    inducing an immunological response and autoantibody produc-

    tion against endogenous angiotensin-I or angiotensin-II.63

    PMD3117 consists of a 12-amino-acid analog of angiotensin-

    I, which is coupled to the carrier, keyhole limpet hemocyanin,

    and adsorbed onto the adjuvant, aluminium hydroxide.64 Pre-

    clinical experiments documented a reduction in blood pressure.

    However, a subsequent phase II clinical trial failed to show a

    similar blood pressure response in humans, although immuno-

    logic activity was documented and there was biochemical evi-

    dence of RAAS inhibition.64 It was postulated that higher titers

    might be required to induce a sustained antihypertensive effect.

    Modifications to the vaccine with a change in its adjuvant have

    been made and additional clinical trials are underway.63

    A second vaccine recently tested in humans is Cyt006-

    AngQb, which targets angiotensin-II.65,66 It is composed of

    an angiotensin-II-derived peptide conjugated to recombinant

    virus-like particles originating from the RNA-phage Qb. In

    phase I trials, the vaccine was well tolerated and generated a

    strong immunogenic response to angiotensin-II. In a small,

    randomized placebo-controlled phase IIa trial of 72 patients

    with mild-to-moderate hypertension, Cyt006-AngQb was well

    tolerated, with no serious side effects and no safety issues.

    After a single dose of the vaccine, all patients generated an

    immune response to angiotensin-II. Following second and third

    injections at 4 and 12 weeks, respectively, the half-life approxi-

    mated 4 months, which supports a treatment regimen of a fewtimes per year. At the 300 mg dose, there were average reduc-

    tions of 4 to 9 mm Hg in daytime diastolic and systolic blood

    pressures and 13 to 25 mm Hg decrease in the early morning

    blood pressure surge, compatible with attenuation of RAAS sti-

    mulation. The effect on early morning blood pressure was

    unexpected and may be unique to the vaccine, due to the slow,

    steady rise in antibody production compared to the peaks and

    troughs of oral agents. Although these initial results are promis-

    ing, they reflected secondary analysis of a very small number

    of patients. There also remain unanswered safety concerns.67

    In regulating fluid and electrolyte balance, RAAS has benefi-

    cial physiologic effects that aid the bodys ability to maintainhemodynamic homeostasis in the setting of various stresses.

    Importantly, its rapid activation is vital in situations such as

    shock, trauma, and severe dehydration. Although arguably,

    antihypertensive medications that inhibit RAAS would also

    theoretically blunt the bodys response to such insults,

    vaccine-induced circulating antibodies to angiotensin-II have

    much longer half-lives, as long as 17 weeks, and cannot be eas-

    ily reversed by mere withdrawal of therapy, as is the case with

    oral medications.67 In addition, even in the setting of milder

    degrees of salt and volume depletion, inhibiting RAAS could

    lead to dangerous levels of hyperkalemia and renal failure.67

    There are also theoretical concerns regarding the triggeringof autoimmune disease by repeated injections of an endogen-

    ous peptide linked to virus-like particles. Furthermore, much

    larger clinical trials of longer duration are needed to confirm

    the antihypertensive effects and safety as well as whether there

    are associated end-organ benefits to vaccination.

    Treatment of Obstructive Sleep Apnea

    Obstructive sleep apnea (OSA) is a strong, independent predic-

    tor of the development of hypertension and the severity of both

    disorders are directly related.6,68-71 Although not fully eluci-

    dated, the relationship between sleep-disordered breathing and

    elevated blood pressure is thought to occur via various neuro-

    hormonal mechanisms. Periodic apnea with hypoxemia and

    hypercapnia cause surges of sympathetic nervous system acti-

    vation and parasympathetic withdrawal, which repetitively

    increase blood pressure.69,71 Blood pressures as high as 240/

    130 mm Hg have been recorded in patients at the end of apneic

    spells.71,72 Obstructive sleep apnea is also associated with

    aldosterone excess, which increases oxidative stress and

    inflammation.69,73 Obesity itself, which is commonly associ-

    ated with OSA, appears to independently activate the sympa-

    thetic nervous system and stimulate RAAS.69,74

    The most effective treatment for OSA is continuous positive

    airway pressure (CPAP) administered via nasal mask.73

    262 Journal of Cardiovascular Pharmacology and Therapeutics 15(3)

    262

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    8/12

    Continuous positive airway pressure therapy results in acute

    reductions in blood pressure and sympathetic activity during

    the sleep cycle.71,72 Data supporting long-term benefits of

    CPAP in hypertensive patients are less definitive, primarily

    because there are relatively few well-designed, longitudinal-

    controlled trials.71,73 Many studies were of short follow-up

    duration and failed to include true placebo-control groups,rather than sham CPAP (application of the nasal mask without

    concomitant pressure) which was subsequently shown to

    increase blood pressure.73,75,76 Trial end points were often spot

    blood pressure measurements taken at variable times of day,

    rather than 24-hour ambulatory blood pressure recordings.71,73 In addition, many of the study patients were either normo-

    tensive or had well-controlled blood pressure at the beginning

    of the study.71,73 In such patients, net reductions of*2 mm Hg

    have been observed, which are modest but statistically signifi-

    cant.71,73 Few studies have exclusively studied patients with

    hypertension; one such study reported a 10 mm Hg decrease

    in systolic and diastolic blood pressures and was characterizedby longer CPAP use and more severe OSA.77 In an observa-

    tional study of refractory hypertensive patients with OSA,

    CPAP treatment for 2 months was associated with an 11 mm

    Hg reduction in 24-hour systolic blood pressure, with a decline

    in nocturnal systolic blood pressure of 14 mm Hg.78

    The existing data suggest modest and heterogeneous blood

    pressure responses to CPAP that are patient specific. The most

    benefit may be derived by individuals in whom (a) OSA is

    more severe (particularly those with prominent complaints

    of daytime somnolence); (b) blood pressure is harder to con-

    trol; and (c) CPAP compliance is high.71,73 However, addi-

    tional well-designed, longer term randomized trials areneeded to further define the role of OSA therapy in reducing

    the risk of developing hypertension and treating patients with

    hypertension.

    Baroreflex Stimulation

    It has long been known that the carotid sinus baroreceptor

    reflex plays a significant role in blood pressure homeostasis

    and modulation of autonomic tone. Arterial stretch receptors

    in the carotid sinuses and aortic arch are exquisitely sensitive

    to arterial stretch. As blood pressure increases, baroreceptor

    activity rises in proportion to the rate of change in arterial pres-

    sure. Impulses are propagated to the central nervous system,

    which results in increased parasympathetic tone in the heart

    and reduced sympathetic activation of the heart, kidneys, and

    peripheral vasculature, causing blood pressure to fall.

    The concept of carotid nerve stimulation arose in the 1950s

    and 1960s.79 It originated from the notion that artificially

    increasing nerve activity from the carotid sinus to the brain

    stem would be interpreted as a rise in arterial blood pressure

    and lead to neural signaling that reduced blood pressure and

    heart rate. Several initial devices using carotid sinus electrical

    stimulation were tested in animals and humans but faced

    numerous technical problems, including uncomfortable extra-

    neous muscle and nerve stimulation, nerve damage, and

    difficulties with communication between an external antenna

    and the implanted receiver. Recent technological advances can

    theoretically overcome many of these limitations.

    The newest carotid sinus stimulator device now under-

    going clinical trials is the Rheos System (CVRx, Mineapolis,

    Minnesota).71,79,80 It received an investigational device exemp-

    tion from the FDA in 2006. Electrodes are placed on both car-otid arteries and the leads are attached to a pulse generator that

    is implanted subcutaneously near the clavicle. Surgical implan-

    tation under general anesthesia is required. The device is fully

    programmable, which facilitates individualized parameter set-

    tings. It was first tested in a small group of 11 patients under-

    going elective carotid surgery as a proof-of-concept study.

    The device resulted in significant reductions in blood pressure

    (18 + 26 mm Hg systolic and 8 + 12 mm Hg diastolic) in a

    voltage-dependent manner. Maximal blood pressure reductions

    were seen using 4.4 + 1.2 volts (23 + 24 mm Hg systolic and

    16 + 10 mm Hg diastolic). Ongoing trials sponsored by the

    manufacturer80,81

    include the device-based therapy in hyper-tension trial (DEBuT-HT), device-based therapy in hyperten-

    sion extension trial (DEBuT-HET), and the Rheos pivotal

    trial. Device-based therapy in hypertension trial is a multicen-

    ter European study examining the safety and efficacy of Rheos

    in 45 patients with refractory hypertension. Results of a sub-

    study of 21 DEBuT-HT patients have recently been reported,

    which investigated whether chronic baroreceptor stimulation

    resulted in changes in cardiac autonomic regulation, as

    reflected by heart rate variability and heart rate turbulence, and

    whether there was a corresponding effect on blood pressure.82

    Measurements were made at 1 month after device implantation

    with the stimulator turned off and compared with measure-ments made after 3 months of activated electrical stimulator

    therapy. There were significant declines in office blood pres-

    sures (185 + 31/109 + 24 mm Hg to 154 + 23/95 +

    16 mm Hg), which were associated with significant changes

    in heart rate variability and heart rate turbulence, supporting

    decreased sympathetic and increased parasympathetic activity

    in actively treated patients. The main results of the safety stud-

    ies, DEBuT-HT, and the related longer term follow-up study,

    DEBuT-HT, are yet to be published. Also in progress is the

    Rheos Pivotal Trial which is a randomized, double-blind trial

    of 300 patients aiming to demonstrate clinically significant,

    sustained reductions in office systolic blood pressures up to 1

    year. Although the initial data were promising, the results of

    these ongoing studies will be required to better define the clin-

    ical role of baroreceptor stimulation.

    Renal Nerve Ablation

    From both preclinical and human studies, it has long been known

    that renal sympathetic innervation plays an important pathophy-

    siologic role in initiating and maintaining hypertension.83,84

    Renal efferent nerve endings are found in the renal arteries, jux-

    taglomerular apparatus, and renal tubules. Activation of the renal

    nerves stimulates renin release, promotes sodium reabsorption,

    and impairs renal perfusion. The renal sensory afferent nerves

    Wu and Gerstenblith 263

    263

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    9/12

    provide feedback regulation to the central nervous system,

    specifically, the posterior hypothalamus, and thus directly con-

    trol the level of sympathetic outflow to not only the kidneys but

    also the heart and peripheral vasculature.

    The potential therapeutic benefits of renal denervation on

    blood pressure control were first observed from preclinical

    experiments.83 The purpose of these studies using animal mod-els was originally to examine the pathophysiologic mechan-

    isms by which the renal efferent and afferent nerves

    contribute to normal and pathologic functioning of the kidneys

    and other organs. It was generally noted in multiple models of

    hypertension that bilateral renal denervation averted the devel-

    opment of hypertension or significantly reduced its magni-

    tude.85,86 This led to efforts to modulate renal innervations

    surgically in patients to control blood pressure. Initial surgical

    efforts involved surgical nephrectomy; thoracolumbar, abdom-

    inal, or pelvic sympathectomy was plaqued by considerable

    morbidity and mortality.83,84 Moreover, such approaches could

    not guarantee uniform efficacy in functionally eliminatingrenal innervation and consequently, the effects on blood pres-

    sure control were erratic.83 However, selective renal denerva-

    tion remains an attractive therapeutic target. Recently, a

    percutaneous, catheter-based approach was developed and

    tested in a proof-of-concept and safety study in 45 patients with

    resistant hypertension.84 A catheter (Symplicity by Ardian Inc,

    Palo Alto, California) was advanced into each renal artery via

    the femoral approach and radiofrequency ablation was per-

    formed at 6 separate sites within each artery. The median pro-

    cedure time was 38 minutes. One patient had a renal artery

    dissection upon placement of the catheter, prior to radiofre-

    quency treatment, and underwent successful renal stenting andwas withdrawn from the renal denervation protocol. There

    were no other renovascular complications for 6 months post-

    procedure and glomerular filtration rates were stable. Limited

    efficacy data suggest long-term, persistent reductions of office

    blood pressures in the 9 patients reaching the 12-month follow-

    up at the time of the studys publication: systolic/diastolic

    reductions of 14/10 + 4/3 mm Hg; 21/10 + 7/4;

    22/11 + 10/5; 24/11 + 9/5; and27/17 + 16/11

    at 1, 3, 6, 9, and 12 months postprocedure, respectively. Nota-

    bly, the study documented a 47% reduction in postprocedure

    noradrenaline spillover from the kidneys into the circulation,

    supporting the effective denervation of the efferent renal sym-

    pathetic nerves by the technique.

    Although this initial report is promising, there are a number

    of limitations and unanswered questions.84,87 Large rando-

    mized trials that include a control group and which better stan-

    dardize or control for concomitant medical therapy are needed.

    In addition, not all patients in the study responded to the ther-

    apy and this may be due to the variable causes of resistant

    hypertension which were not well-defined or investigated.

    Hence, a better defined target group of study patients is needed.

    The effects on ambulatory blood pressure rather than office

    blood pressure may better characterize the true benefits of the

    therapy. Finally, this approach is not likely to be recommended

    as a first-line therapy in the management of hypertension but

    reserved for specific patient subgroups, yet to be defined, with

    resistant hypertension.

    Summary

    Hypertension poses a growing public health dilemma of epi-

    demic proportions. It is expected to affect 1.56 billion individ-uals worldwide by year 2025, both in developed and

    developing countries.88 Although there are a number of effec-

    tive treatments, the majority of patients do not achieve target

    blood pressure for a variety of reasons, including insufficient

    medication, true drug resistance, and noncompliance. There

    remains a significant need to develop and thoroughly test new

    agents and approaches to better control hypertension, in the

    form of drugs that target multiple components of the pathways,

    which control blood pressure and/or via novel drug delivery

    such as vaccination.

    Declaration of Conflicting Interests

    The authors declared no conflicts of interest with respect to the author-

    ship and/or publication of this article.

    Financial Disclosure/Funding

    The authors disclosed receipt of the following financial support for the

    research and/or authorship of this article: Dr Wu is supported by the

    Donald W. Reynolds Cardiovascular Research Center at Johns Hop-

    kins University.

    References

    1. American Heart Association. Heart disease and stroke statistics2009 update.

    2. Fields LE, Burt VL, Cutler JA, Hughes J, Roccella EJ, Sorlie P.

    The burden of adult hypertension in the United States 1999 to

    2000: a rising tide. Hypertension. 2004;44(4):398-404.

    3. Yechiam O, Charles FD, Jeffery PH, Margaret C, Sarah Y. Trends

    in hypertension prevalence, awareness, treatment, and control in

    older U.S. adults: data from the National Health and Nutrition

    Examination Survey 1988 to 2004. J Am Geriatr Soc. 2007;

    55(7):1056-1065.

    4. Gu Q, Dillon CF, Burt VL, Gillum RF. Association of hypertension

    treatment and control with all-cause and cardiovascular disease

    mortality among US adults with hypertension. Am J Hypertens.

    2010;23(1):38-45.

    5. Black HR. Triple fixed-dose combination therapy: back to the past.

    Hypertension. 2009;54(1):19-22.

    6. Chobanian AV, Bakris GL, Black HR, et al. Seventh report of the

    Joint National Committee on prevention, detection, evaluation, and

    treatment of high blood pressure. Hypertension. 2003;42(6):

    1206-1252.

    7. Munzel T, Gori T. Nebivolol: the somewhat-different beta-

    adrenergic receptor blocker. J Am Coll Cardiol. 2009;54(16):

    1491-1499.

    8. Manrique C, Giles TD, Ferdinand KC, Sowers JR. Realities of

    newer beta-blockers for the management of hypertension. J Clin

    Hypertens (Greenwich). 2009;11(7):369-375.

    264 Journal of Cardiovascular Pharmacology and Therapeutics 15(3)

    264

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    10/12

    9. Nebivolol (Bystolic) for hypertension. Med Lett Drugs Ther.

    2008;50(1281):17-19.

    10. Mason RP, Giles TD, Sowers JR. Evolving mechanisms of action

    of beta blockers: focus on nebivolol. J Cardiovasc Pharmacol.

    2009;54(2):123-128.

    11. Flather MD, Shibata MC, Coats AJ, et al. Randomized trial to

    determine the effect of nebivolol on mortality and cardiovascularhospital admission in elderly patients with heart failure

    (SENIORS). Eur Heart J. 2005;26(3):215-225.

    12. Weiss RJ, Weber MA, Carr AA, Sullivan WA. A randomized,

    double-blind, placebo-controlled parallel-group study to assess

    the efficacy and safety of nebivolol, a novel beta-blocker, in

    patients with mild to moderate hypertension. J Clin Hypertens

    (Greenwich). 2007;9(9):667-676.

    13. Saunders E, Smith WB, DeSalvo KB, Sullivan WA. The efficacy

    and tolerability of nebivolol in hypertensive African American

    patients. J Clin Hypertens (Greenwich). 2007;9(11):866-875.

    14. Van Nueten L, Taylor FR, Robertson JI. Nebivolol vs atenolol and

    placebo in essential hypertension: a double-blind randomizedtrial. J Hum Hypertens. 1998;12(2):135-140.

    15. Czuriga I, Riecansky I, Bodnar J, et al, For The NEBIS Investiga-

    tors; NEBIS Investigators Group. Comparison of the new cardio-

    selective beta-blocker nebivolol with bisoprolol in hypertension:

    the Nebivolol, Bisoprolol Multicenter Study (NEBIS). Cardio-

    vasc Drugs Ther. 2003;17(3):257-263.

    16. Van Nueten L, Schelling A, Vertommen C, Dupont AG,

    Robertson JI. Nebivolol vs enalapril in the treatment of essential

    hypertension: a double-blind randomized trial. J Hum Hypertens.

    1997;11(12):813-819.

    17. Van Bortel LM, Bulpitt CJ, Fici F. Quality of life and antihyper-

    tensive effect with nebivolol and losartan. Am J Hypertens. 2005;

    18(8):1060-1066.

    18. Rosei EA, Rizzoni D, Comini S, Boari G. Evaluation of the effi-

    cacy and tolerability of nebivolol versus lisinopril in the treatment

    of essential arterial hypertension: a randomized, multicentre,

    double-blind study. Blood Press Suppl. 2003:1:30-35.

    19. Van Nueten L, Lacourciere Y, Vyssoulis G, et al. Nebivolol ver-

    sus nifedipine in the treatment of essential hypertension: a double-

    blind, randomized, comparative trial. Am J Ther. 1998;5(4):

    237-243.

    20. Papademetriou V. Comparison of Nebivolol monotherapy versus

    Nebivolol in combination with other antihypertensive therapies

    for the treatment of hypertension. Am J Cardiol. 2009;103(2):

    273-278.

    21. Bangalore S, Sawhney S, Messerli FH. Relation of beta-blocker-

    induced heart rate lowering and cardioprotection in hypertension.

    J Am Coll Cardiol. 2008;52(18):1482-1489.

    22. Lavie CJ, Messerli FH, Milani RV. Beta-blockers as first-line

    antihypertensive therapy the crumbling continues. J Am Coll Car-

    diol. 2009;54(13):1162-1164.

    23. Dockery BK, Bisognano JD. Direct renin inhibition: an analy-

    sis of possible benefits. Curr Hypertens Rep. 2008;10(4):

    313-318.

    24. Yusuf S, Teo KK, Pogue J, et al. Telmisartan, ramipril, or both in

    patients at high risk for vascular events. N Engl J Med. 2008;

    358(15):1547-1559.

    25. Rongen GA, Lenders JW, Smits P, Thien T. Clinical pharmacoki-

    netics and efficacy of renin inhibitors. Clin Pharmacokinet. 1995;

    29(1):6-14.

    26. Staessen JA, Li Y, Richart T. Oral renin inhibitors. Lancet. 2006;

    368(9545):1449-1456.

    27. Rahuel J, Rasetti V, Maibaum J, et al. Structure-based drug

    design: the discovery of novel nonpeptide orally active inhibitorsof human renin. Chem Biol. 2000;7(7):493-504.

    28. Stanton A, Jensen C, Nussberger J, OBrien E. Blood pressure

    lowering in essential hypertension with an oral renin inhibitor,

    aliskiren. Hypertension. 2003;42(6):1137-1143.

    29. Vaidyanathan S, Camenisch G, Schuetz H, et al. Pharmacoki-

    netics of the oral direct renin inhibitor aliskiren in combination

    with digoxin, atorvastatin, and ketoconazole in healthy subjects:

    the role of P-glycoprotein in the disposition of aliskiren. J Clin

    Pharmacol. 2008;48(11):1323-1338.

    30. Aliskiren for hypertension in adults. Drug Ther Bull. 2008;46(10):

    73-76.

    31. Brown MJ. Aliskiren. Circulation. 2008;118(7):773-784.32. Gradman AH, Schmieder RE, Lins RL, Nussberger J, Chiang Y,

    Bedigian MP. Aliskiren, a novel orally effective renin inhibitor,

    provides dose-dependent antihypertensive efficacy and placebo-

    like tolerability in hypertensive patients. Circulation. 2005;

    111(8):1012-1018.

    33. Schmieder RE, Philipp T, Guerediaga J, et al. Long-term antihy-

    pertensive efficacy and safety of the oral direct renin inhibitor

    aliskiren: a 12-month randomized, double-blind comparator trial

    with hydrochlorothiazide. Circulation. 2009;119(3):417-425.

    34. Gradman AH, Kad R. Renin inhibition in hypertension. J Am Coll

    Cardiol. 2008;51(5):519-528.

    35. Solomon SD, Appelbaum E, Manning WJ, et al. Effect of the

    direct renin inhibitor aliskiren, the angiotensin receptor blocker

    losartan, or both on left ventricular mass in patients with hyper-

    tension and left ventricular hypertrophy. Circulation. 2009;

    119(4):530-537.

    36. McInnes GT. Renin inhibition: the holy grail of renin-angiotensin

    system blockade? J Hum Hypertens. 2007;21(10):766-769.

    37. Jensen C, Herold P, Brunner HR. Aliskiren: the first renin inhibi-

    tor for clinical treatment. Nat Rev Drug Discov. 2008;7(5):

    399-410.

    38. Parving HH, Brenner BM, McMurray JJ, et al. Aliskiren Trial in

    Type 2 Diabetes Using Cardio-Renal Endpoints (ALTITUDE):

    rationale and study design. Nephrol Dial Transplant. 2009;

    24(5):1663-1671.

    39. Yanagisawa M, Kurihara H, Kimura S, et al. A novel potent vaso-

    constrictor peptide produced by vascular endothelial cells.

    Nature. 1988;332(6163):411-415.

    40. Prasad VS, Palaniswamy C, Frishman WH. Endothelin as a clin-

    ical target in the treatment of systemic hypertension. Cardiol Rev.

    2009;17(4):181-191.

    41. Ge Y, Bagnall A, Stricklett PK, Webb D, Kotelevtsev Y,

    Kohan DE. Combined knockout of collecting duct endothelin A

    and B receptors causes hypertension and sodium retention. Am

    J Physiol Renal Physiol. 2008;295(6):F1635-F1640.

    42. Krum H, Viskoper RJ, Lacourciere Y, Budde M, Charlon V. The

    effect of an endothelin-receptor antagonist, bosentan, on blood

    Wu and Gerstenblith 265

    265

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    11/12

    pressure in patients with essential hypertension. Bosentan

    Hypertension Investigators. N Engl J Med. 1998;338(12):784-790.

    43. Enseleit F, Luscher TF, Ruschitzka F. Darusentan: a new perspec-

    tive for treatment of resistant hypertension? Expert Opin Investig

    Drugs. 2008;17(8):1255-1263.

    44. Weber MA, Black H, Bakris G, et al. A selective endothelin-

    receptor antagonist to reduce blood pressure in patients withtreatment-resistant hypertension: a randomized, double-blind,

    placebo-controlled trial. Lancet. 2009;374(9699):1423-1431.

    45. Gilead Sciences. Second Pivotal Phase III Study of Gileads Dar-

    usentan for Resistant Hypertension Misses Primary Endpoints

    [press release]. http://www.gilead.com/pr_1365747. Accessed

    February 25, 2010.

    46. Dive V, Chang CF, Yiotakis A, Sturrock ED. Inhibition of zinc

    metallopeptidases in cardiovascular diseasefrom unity to tri-

    nity, or duality? Curr Pharm Des. 2009;15(31):3606-3621.

    47. Bevan EG, Connell JM, Doyle J, et al. Candoxatril, a neutral

    endopeptidase inhibitor: efficacy and tolerability in essential

    hypertension. J Hypertens. 1992;10(7):607-613.48. Singer DR, Markandu ND, Buckley MG, Miller MA,

    Sagnella GA, MacGregor GA. Dietary sodium and inhibition of

    neutral endopeptidase 24.11 in essential hypertension. Hyperten-

    sion. 1991;18(6):798-804.

    49. Daull P, Jeng AY, Battistini B. Towards triple vasopeptidase inhi-

    bitors for the treatment of cardiovascular diseases. J Cardiovasc

    Pharmacol. 2007;50(3):247-256.

    50. Kostis JB, Packer M, Black HR, Schmieder R, Henry D, Levy E.

    Omapatrilat and enalapril in patients with hypertension: the Oma-

    patrilat Cardiovascular Treatment vs Enalapril (OCTAVE) trial.

    Am J Hypertens. 2004;17(2):103-111.

    51. Trippodo NC, Robl JA, Asaad MM, Fox M, Panchal BC,

    Schaeffer TR. Effects of omapatrilat in low, normal, and high

    renin experimental hypertension. Am J Hypertens. 1998;11(3 pt

    1):363-372.

    52. Messerli FH, Nussberger J. Vasopeptidase inhibition and angio-

    oedema. Lancet. 2000;356(9230):608-609.

    53. Tabrizchi R. Ilepatril (AVE-7688), a vasopeptidase inhibitor for

    the treatment of hypertension. Curr Opin Investig Drugs. 2008;

    9(3):301-309.

    54. Gu J, Noe A, Chandra P, et al. Pharmacokinetics and pharmaco-

    dynamics of LCZ696, a novel dual-acting angiotensin receptor-

    neprilysin inhibitor (ARNi). J Clin Pharmacol. 2010;50(4):

    401-414.

    55. Dickstein K, De Voogd HJ, Miric MP, et al. Effect of single doses

    of SLV306, an inhibitor of both neutral endopeptidase and

    endothelin-converting enzyme, on pulmonary pressures in con-

    gestive heart failure. Am J Cardiol. 2004;94(2):237-239.

    56. Heiss G, Wallace R, Anderson GL, et al. Health risks and benefits

    3 years after stopping randomized treatment with estrogen and

    progestin. JAMA. 2008;299(9):1036-1045.

    57. Drospirenone in HRT? Drug Ther Bull. 2009;47(4):41-44.

    58. Krattenmacher R. Drospirenone: pharmacology and pharmacoki-

    netics of a unique progestogen. Contraception. 2000;62(1):

    29-38.

    59. White WB, Hanes V, Chauhan V, Pitt B. Effects of a new hor-

    mone therapy, drospirenone and 17-beta-estradiol, in

    postmenopausal women with hypertension. Hypertension. 2006;

    48(2):246-253.

    60. White WB, Pitt B, Preston RA, Hanes V. Antihypertensive effects

    of drospirenone with 17beta-estradiol, a novel hormone treatment

    in postmenopausal women with stage 1 hypertension. Circulation.

    2005;112(13):1979-1984.

    61. Sica DA. Drospirenone: an antihypertensive in waiting. Hyperten-sion. 2006;48(2):205-206.

    62. Perreault S, Lamarre D, Blais L, et al. Persistence with treatment

    in newly treated middle-aged patients with essential hypertension.

    Ann Pharmacother. 2005;39(9):1401-1408.

    63. Brown MJ. Success and failure of vaccines against renin-

    angiotensin system components. Nat Rev Cardiol. 2009;6(10):

    639-647.

    64. Brown MJ, Coltart J, Gunewardena K, Ritter JM, Auton TR,

    Glover JF. Randomized double-blind placebo-controlled study

    of an angiotensin immunotherapeutic vaccine (PMD3117) in

    hypertensive subjects. Clin Sci (Lond). 2004;107(2):167-173.

    65. Ambuhl PM, Tissot AC, Fulurija A, et al. A vaccine for hyperten-sion based on virus-like particles: preclinical efficacy and phase I

    safety and immunogenicity. J Hypertens. 2007;25(1):63-72.

    66. Tissot AC, Maurer P, Nussberger J, et al. Effect of immunization

    against angiotensin II with CYT006-AngQb on ambulatory blood

    pressure: a double-blind, randomized, placebo-controlled phase

    IIa study. Lancet. 2008;371(9615):821-827.

    67. Samuelsson O, Herlitz H. Vaccination against high blood pres-

    sure: a new strategy. Lancet. 2008;371(9615):788-789.

    68. Nieto FJ, Young TB, Lind BK, et al. Association of sleep-

    disordered breathing, sleep apnea, and hypertension in a large

    community-based study. Sleep Heart Health Study. JAMA.

    2000;283(14):1829-1836.

    69. Pimenta E, Calhoun DA, Oparil S. Sleep apnea, aldosterone, and

    resistant hypertension. Prog Cardiovasc Dis. 2009;51(5):

    371-380.

    70. Peppard PE, Young T, Palta M, Skatrud J. Prospective study of

    the association between sleep-disordered breathing and hyperten-

    sion. N Engl J Med. 2000;342(19):1378-1384.

    71. Somers VK, White DP, Amin R, et al. Sleep apnea and cardiovas-

    cular disease: an American Heart Association/American College

    of Cardiology Foundation Scientific Statement from the Ameri-

    can Heart Association Council for High Blood Pressure Research

    Professional Education Committee, Council on Clinical Cardiol-

    ogy, Stroke Council, and Council on Cardiovascular Nursing.

    J Am Coll Cardiol. 2008;52(8):686-717.

    72. Somers VK, Dyken ME, Clary MP, Abboud FM. Sympathetic

    neural mechanisms in obstructive sleep apnea. J Clin Invest.

    1995;96(4):1897-1904.

    73. Bradley TD, Floras JS. Obstructive sleep apnoea and its cardio-

    vascular consequences. Lancet. 2009;373(9657):82-93.

    74. Esler M, Straznicky N, Eikelis N, Masuo K, Lambert G,

    Lambert E. Mechanisms of sympathetic activation in obesity-

    related hypertension. Hypertension. 2006;48(5):787-796.

    75. Norman D, Loredo JS, Nelesen RA, et al. Effects of continuous

    positive airway pressure versus supplemental oxygen on

    24-hour ambulatory blood pressure. Hypertension. 2006;47(5):

    840-845.

    266 Journal of Cardiovascular Pharmacology and Therapeutics 15(3)

    266

    at HINARI on November 3, 2012cpt.sagepub.comDownloaded from

    http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/http://cpt.sagepub.com/
  • 7/30/2019 NUEVOS MEDICAMENTOS ANTIHIPERTENSIVOS

    12/12

    76. Pepperell JC, Ramdassingh-Dow S, Crosthwaite N, et al. Ambu-

    latory blood pressure after therapeutic and subtherapeutic nasal

    continuous positive airway pressure for obstructive sleep apnoea:

    a randomized parallel trial. Lancet. 2002;359(9302):204-210.

    77. Becker HF, Jerrentrup A, Ploch T, et al. Effect of nasal contin-

    uous positive airway pressure treatment on blood pressure in

    patients with obstructive sleep apnea. Circulation. 2003;107(1):68-73.

    78. Logan AG, Tkacova R, Perlikowski SM, et al. Refractory hyper-

    tension and sleep apnoea: effect of CPAP on blood pressure and

    baroreflex. Eur Respir J. 2003;21(2):241-247.

    79. Doumas M, Guo D, Papademetriou V. Carotid baroreceptor

    stimulation as a therapeutic target in hypertension and other car-

    diovascular conditions. Expert Opin Ther Targets. 2009;13(4):

    413-425.

    80. CVRx. For Medical ProfessionalsProducts. http://www.cvrx.

    com/medpros/index.php?id15. Accessed February 25, 2010.

    81. National Institutes of Health. Device Based Therapy in Hyperten-

    sion Extension Trial (DEBuT-HET). http://clinicaltrials.gov/ct2/show/NCT00710294. Accessed February 25, 2010.

    82. Wustmann K, Kucera JP, Scheffers I, et al. Effects of chronic bar-

    oreceptor stimulation on the autonomic cardiovascular regulation

    in patients with drug-resistant arterial hypertension. Hypertension.

    2009;54(3):530-536.

    83. Schlaich MP, Sobotka PA, Krum H, Whitbourn R, Walton A,

    Esler MD. Renal denervation as a therapeutic approach for hyper-

    tension: novel implications for an old concept. Hypertension.2009;54(6):1195-1201.

    84. Krum H, Schlaich M, Whitbourn R, et al. Catheter-based renal

    sympathetic denervation for resistant hypertension: a multicentre

    safety and proof-of-principle cohort study. Lancet. 2009;

    373(9671):1275-1281.

    85. DiBona GF, Kopp UC. Neural control of renal function. Physiol

    Rev. 1997;77(1):75-197.

    86. DiBona GF. The sympathetic nervous system and hypertension:

    recent developments. Hypertension. 2004;43(2):147-150.

    87. Doumas M, Douma S. Interventional management of resistant

    hypertension. Lancet. 2009;373(9671):1228-1230.

    88. Hypertension: uncontrolled and conquering the world. Lancet.2007;370(9587):539-539.

    Wu and Gerstenblith 267

    267