吲哚布芬(美国胸科协会抗血小板药物指南)

吲哚布芬(美国胸科协会抗血小板药物指南)
吲哚布芬(美国胸科协会抗血小板药物指南)

DOI 10.1378/chest.08-0672

2008;133;199S-233S Chest

Carlo Patrono, Colin Baigent, Jack Hirsh and Gerald Roth

*

Antiplatelet Drugs

ull.html https://www.360docs.net/doc/b711003600.html,/content/133/6_suppl/199S.f and services can be found online on the World Wide Web at: The online version of this article, along with updated information

ISSN:0012-3692

)https://www.360docs.net/doc/b711003600.html,/site/misc/reprints.xhtml (of the copyright holder.

may be reproduced or distributed without the prior written permission Northbrook, IL 60062. All rights reserved. No part of this article or PDF by the American College of Chest Physicians, 3300 Dundee Road, 2008Physicians. It has been published monthly since 1935. Copyright CHEST is the official journal of the American College of Chest

Antiplatelet Drugs*

American College of Chest Physicians

Evidence-Based Clinical Practice Guidelines

(8th Edition)

Carlo Patrono,MD;Colin Baigent,MD;Jack Hirsh,MD,FCCP;

and Gerald Roth,MD

This article about currently available antiplatelet drugs is part of the Antithrombotic and Thrombo-lytic Therapy:American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition).It describes the mechanism of action,pharmacokinetics,and pharmacodynamics of aspirin,reversible cyclooxygenase inhibitors,thienopyridines,and integrin?IIb?3receptor antago-nists.The relationships among dose,efficacy,and safety are thoroughly discussed,with a mechanistic overview of randomized clinical trials.The article does not provide specific management recommen-dations;however,it does highlight important practical aspects related to antiplatelet therapy, including the optimal dose of aspirin,the variable balance of benefits and hazards in different clinical settings,and the issue of interindividual variability in response to antiplatelet drugs.

(CHEST2008;133:199S–233S)

Key words:abciximab;antiplatelet drugs;aspirin;clopidogrel;dipyridamole;eptifibatide;platelet pharmacology;resistance; ticlopidine;tirofiban

Abbreviations:ACE?angiotensin-converting enzyme;ADP?adenosine diphosphate;AMP?adenosine monophos-phate;ATT?Antithrombotic Trialists;CAPRIE?Clopidogrel vs Aspirin in Patients at Risk of Ischemic Events; CHD?coronary heart disease;CI?confidence interval;COMMIT?Clopidogrel and Metoprolol Myocardial Infarction Trial;COX?cyclooxygenase;CURE?Clopidogrel in Unstable Angina to Prevent Recurrent Events;EPIC?Evaluation of 7E3for the Prevention of Ischemic Complications;ESPS?European Stroke Prevention Study;ESPRIT?European Stroke Prevention Reversible Ischemia Trial;FDA?Food and Drug Administration;GP?glycoprotein;INR?international normalized ratio;MI?myocardial infarction;NSAID?nonsteroidal antiinflammatory drug;OR?odds ratio; PCI?percutaneous coronary intervention;PE?pulmonary embolism;PG?prostaglandin;PTCA?percutaneous trans-luminal coronary angioplasty;RR?rate ratio;TIA?transient ischemic attack;TX?thromboxane;TTP?thrombotic thrombocytopenic purpura

P latelets are vital components of normal hemosta-sis and key participants in atherothrombosis by virtue of their capacity to adhere to injured blood vessels and to accumulate at sites of injury.1Al-though platelet adhesion and activation can be viewed as a physiologic repair response to the sud-den fissuring or rupture of an atherosclerotic plaque, uncontrolled progression of such a process through a series of self-sustaining amplification loops can lead to intraluminal thrombus formation,vascular occlu-sion,and transient ischemia or infarction.Currently available antiplatelet drugs interfere with some steps in the activation process,including adhesion,release, and/or aggregation,1and have a measurable impact on the risk of arterial thrombosis that cannot be dissociated from an increased risk of bleeding.2

In discussing antiplatelet drugs,it is important to appreciate that approximately1011platelets are pro-duced each day under physiologic circumstances,a level of production that can increase up to10-fold at

*From the Catholic University School of Medicine(Dr.Patrono), Rome,Italy;Clinical Trial Service Unit(Dr.Baigent),University of Oxford,Oxford,UK;Hamilton Civic Hospitals(Dr.Hirsh), Henderson Research Centre,Hamilton,ON,Canada;and Seattle VA Medical Center(Dr.Roth),Seattle,WA.

Dr.Patrono was supported in part by a grant from the European Commission FP6(LSHM-CT-2004-005033).

Manauscript accepted December20,2007.

Reproduction of this article is prohibited without written permission from the American College of Chest Physicians(www.chestjournal. org/misc/reprints.shtml).

Correspondence to:Carlo Patrono,MD,Catholic University School of Medicine,Largo F.Vito1,00168Rome,Italy;e-mail: carlo.patrono@rm.unicatt.it

DOI:10.1378/chest.08-0672Supplement

ANTITHROMBOTIC AND THROMBOLYTIC THERAPY8TH ED:ACCP GUIDELINES https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT199S

times of increased need.3Platelets are anucleate blood cells that form by fragmentation of megakaryo-cyte cytoplasm and have a maximum circulating life span of about10days in humans.3Platelets provide a circulating source of chemokines,cytokines,and growth factors,which are preformed and packaged in storage granules.Moreover,activated platelets can synthesize prostanoids(primarily,thromboxane[TX] A2)from arachidonic acid released from membrane phospholipids through rapid coordinated activation of phospholipase(s),cyclooxygenase(COX)-1and TX synthase(Fig1).Newly formed platelets also express the inducible isoforms of COX(COX-2)and prosta-glandin(PG)E synthase,and this phenomenon is markedly amplified in association with accelerated platelet regeneration.4Although activated platelets are not thought to synthesize proteins de novo,they can translate constitutive messenger RNAs into pro-teins,including interleukin-1?,over several hours.5 Thus,platelets may play previously unrecognized roles in inflammation and vascular injury,and anti-platelet strategies may be expected to affect platelet-derived protein signals for inflammatory and/or pro-liferative responses.1

Negative modulation of platelet adhesion and aggre-gation is exerted by a variety of physiologic mecha-nisms,including endothelium-derived prostacyclin (PGI2),nitric oxide,CD39/ecto-ADPase,and platelet endothelial cell adhesion molecule-1.Some drugs may interfere with these regulatory pathways,as exempli-fied by the dose-dependent inhibition of PGI2produc-tion by aspirin and other COX inhibitors.2

2.0Aspirin and Other COX Inhibitors Aspirin has been thoroughly evaluated as an anti-platelet drug6and was found to prevent vascular death by approximately15%and nonfatal vascular events by about30%in a metaanalysis of?100 randomized trials in high-risk patients.7

2.1Mechanism of Action of Aspirin

The best characterized mechanism of action of the drug is related to its capacity to inactivate permanently the COX activity of prostaglandin H-synthase-1and-2 (also referred to as COX-1and COX-2).8–12These isozymes catalyze the first committed step in prosta-noid biosynthesis(ie,the conversion of arachidonic acid to PGH2)[Fig1].PGH2is the immediate precursor of PGD2,PGE2,PGF2?,PGI2,and TXA2. COX-1and COX-2are homodimers of a?72kd monomeric unit.Each dimer has three independent folding units:an epidermal growth factor-like do-main;a membrane-binding domain;and an

enzy-

Figure1.Arachidonic acid metabolism and mechanism of action of aspirin.Arachidonic acid,a

20-carbon fatty acid containing four double bonds,is liberated from the sn2position in membrane

phospholipids by several forms of phospholipase,which are activated by diverse stimuli.Arachidonic

acid is converted by cytosolic PGH synthases,which have both COX and hydroperoxidase activity,to

the unstable intermediate PGH2.The synthases are colloquially termed COXs and exist in two forms,

COX-1and COX-2.Low-dose aspirin selectively inhibits COX-1,and high-dose aspirin inhibits both

COX-1and COX-2.PGH2is converted by tissue-specific isomerases to multiple prostanoids.These

bioactive lipids activate specific cell membrane receptors of the superfamily of G-protein-coupled

receptors.DP?PGD2receptor;EP?PGE2receptor;FP?PGF2?receptor;IP?prostacyclin

receptor;TP?TX receptor.

200S Antithrombotic and Thrombolytic Therapy8th Ed:ACCP Guidelines

matic domain.12Within the enzymatic domain,there is the peroxidase catalytic site and a separate,but adjacent site for COX activity at the apex of a narrow, hydrophobic channel.

The molecular mechanism of permanent inactiva-tion of COX activity by aspirin is related to blockade of the COX channel as a consequence of acetylation of a strategically located serine residue(Ser529in the human COX-1,Ser516in the human COX-2) that prevents access of the substrate to the catalytic site of the enzyme.13The hydrophobic environment of the COX channel stabilizes the modified serine side-chain against hydrolysis.13Thus,inhibition of COX-1–dependent platelet function can be achieved with low doses of aspirin given once daily.In con-trast,inhibition of COX-2–dependent pathophysio-logic processes(eg,hyperalgesia and inflammation) requires larger doses of aspirin(probably because acetylation is determined by the oxidative state of the enzyme and is inhibited in cells with high peroxide tone)14and a much shorter dosing interval(because nucleated cells rapidly resynthesize the enzyme).Thus, there is an approximately100-fold variation in daily doses of aspirin when used as an antiinflammatory rather than as an antiplatelet agent.Furthermore, the benefit/risk profile of the drug depends on the dose and indication because its GI toxicity is dose dependent(see below).

Human platelets and vascular endothelial cells process PGH2to produce primarily TXA2and PGI2, respectively.11TXA2induces platelet aggregation and vasoconstriction,whereas PGI2inhibits platelet aggregation and induces vasodilation.11Whereas TXA2is largely a COX-1–derived product(mostly from platelets)and thus highly sensitive to aspirin inhibition,vascular PGI2can derive both from COX-1and,to a greater extent even under physio-logic conditions,from COX-2.16COX-1–dependent PGI2production occurs transiently in response to agonist stimulation(eg,bradykinin)15and is sensitive to aspirin inhibition.COX-2–mediated PGI2produc-tion occurs long term in response to laminar shear stress17and is largely insensitive to aspirin inhibition at conventional antiplatelet doses.This may explain the substantial residual COX-2–dependent PGI2 biosynthesis in vivo at daily doses of aspirin in the range of30to100mg,18despite transient suppres-sion of COX-1–dependent PGI2release.15It is not established that more profound suppression of PGI2 formation by higher doses of aspirin is sufficient to initiate or predispose to thrombosis.However,two lines of evidence suggest that PGI2is thrombopro-tective.The first is the observation that mice lacking the PGI2receptor had increased susceptibility to experimental thrombosis.19The second is the obser-vation of the cardiovascular toxicity associated with COX-2inhibitors20that also supports the concept of PGI2acting as an important mechanism of throm-boresistance in the setting of inadequate inhibition of platelet TXA2biosynthesis.21

2.2Pharmacokinetics

Aspirin is rapidly absorbed in the stomach and upper intestine.Peak plasma levels occur30to40 min after aspirin ingestion,and inhibition of plate-let function is evident by1h.In contrast,it can take up to3to4h to reach peak plasma levels after administration of enteric-coated aspirin.If only enteric-coated tablets are available,and a rapid effect is required,the tablets should be chewed.The oral bioavailability of regular aspirin tablets is ap-proximately40to50%over a wide range of doses.22 A considerably lower bioavailability has been re-ported for enteric-coated tablets and sustained-release,microencapsulated preparations.22Lower bioavailability of some enteric-coated preparations and poor absorption from the higher pH environ-ment of the small intestine may result in inadequate platelet inhibition,particularly in heavier subjects.23 Both a controlled-release formulation15and a trans-dermal patch24with negligible systemic bioavailabil-ity have been developed in an attempt to achieve selective inhibition of platelet TXA2production with-out suppressing systemic PGI2synthesis.The former was used successfully in the Thrombosis Prevention Trial(see below),but it remains unknown whether there is any advantage to the controlled-release formulation vis-a`-vis plain aspirin.

The plasma concentration of aspirin decays with a half-life of15to20min.Despite the rapid clearance of aspirin from the circulation,the platelet-inhibitory effect lasts for the life span of the platelet25because aspirin irreversibly inactivates platelet COX-1.8,9As-pirin also acetylates the enzyme in megakaryocytes before new platelets are released into the circula-tion.10,26–28The mean life span of human platelets is approximately8to10days.Therefore,about10to 12%of circulating platelets are replaced every 24h.29,30However,the recovery of TXA2biosynthe-sis in vivo following prolonged aspirin administration is somewhat faster than predicted by the rate of platelet turnover,18possibly because of the nonlinear relationship between inhibition of platelet COX-1 activity and inhibition of TXA2biosynthesis in vivo31 (Fig2).

2.3Issues Concerning the Antithrombotic Effects of Aspirin

A number of issues related to the clinical efficacy of aspirin continue to be debated.These include the following:(1)the optimal dose of aspirin in order to

https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT201S

maximize efficacy and minimize toxicity;(2)the

suggestion that part of the antithrombotic effect of aspirin is unrelated to inhibition of platelet TXA 2;and (3)the possibility that some patients may be aspirin “resistant.”

2.3.1The Optimal Dose of Aspirin:Well-designed,placebo-controlled randomized trials have shown that aspirin is an effective antithrombotic agent when used long term in doses ranging from 50to 100mg/d,and there is a suggestion that it is effective in doses as low as 30mg/d.6,7Aspirin,75mg/d,was shown to be effective in reducing the risk of acute myocardial infarction (MI)or death in patients with unstable angina 32and chronic stable angina,33as well as in reducing stroke or death in patients with transient cerebral ischemia 34and the risk of postoperative stroke after carotid endarterectomy.35In the Euro-pean Stroke Prevention Study (ESPS)-2,aspirin 25mg bid was effective in reducing the risks of stroke and of the composite outcome stroke or death in patients with prior stroke or transient ischemic attack (TIA).36Moreover,in the European Collaboration on Low-Dose Aspirin in Polycythemia vera Trial,37aspirin,100mg/d,was effective in preventing throm-botic complications in patients with polycythemia vera,despite a higher-than-normal platelet count.

The lowest effective dose of aspirin for these various indications is shown in Table 1.

The clinical effects of different doses of aspirin have been compared directly in a relatively small number of randomized trials.38–43In the United Kingdom TIA study,41no difference in efficacy was found between 300and 1,200mg/d of aspirin (see below).In a study of 3,131patients after a TIA or minor ischemic stroke,aspirin in a dose of 30mg/d was compared with a dose of 283mg/d,and the

Extra Platelet Sources

Platelet Synthesis

TXA 2

TXB 22,3-dinor-TXB 2

& other metabolites

Urine

TXB 2Production In Vivo

Enzymes

Liver H 2O

Calculated Rate of TXB 2

Production In Vivo : 0.1 ng/kg/min

P e r c e n t a g e I n h i b i t i o n o f U r i n a r y 2,3-d i n o r -T X B 2E x c r e t i o n I n V i v o

Percentage Inhibition of Serum

TXB 2Ex Vivo

Pharmacologic Inhibition

Ex Vivo vs In Vivo

TXB 2Production Ex Vivo

100

200

3000

30

60

min

S e r u m T X B 2n g /m l

Time

Whole Blood Clotting at 37°C

Maximal Biosynthetic Capacity 300-400 ng/ml in 1 hr

Figure 2.Maximal capacity of human platelets to synthesize TXB 2,rate of TXB 2production in healthy subjects,and relationship between the inhibition of platelet COX activity and TXB 2biosynthesis in vivo .Left panel:The level of TXB 2production stimulated by endogenous thrombin during whole-blood clotting at 37°C.62Center panel:The metabolic fate of TXA 2in vivo and the calculated rate of its production in healthy subjects on the basis of TXB 2infusions and measurement of its major urinary metabolite.Right panel:The nonlinear relationship between inhibition of serum TXB 2measured ex vivo and the reduction in the excretion of TX metabolite measured in vivo .31Table 1—Vascular Disorders for Which Aspirin Has Been Shown To Be Effective and Lowest Effective Dose

(Section 2.3.1)

Disorder

Lowest Effective Daily Dose,mg

TIA and ischemic stroke *

50Men at high cardiovascular risk 75Hypertension 75Stable angina 75Unstable angina *

75Severe carotid artery stenosis *75Polycythemia vera 100Acute MI

160Acute ischemic stroke *

160

*Higher doses have been tested in other trials and were not found to confer any greater risk reduction.

202S

Antithrombotic and Thrombolytic Therapy 8th Ed:ACCP Guidelines

hazard ratio for the group receiving the lower dose was0.91(95%confidence interval[CI],0.76to 1.09).42The Acetylsalicylic Acid and Carotid Endar-terectomy Trial reported that the risk of stroke,MI, or death within3months of carotid endarterectomy is significantly lower for patients taking81or325 mg/d aspirin than for those taking650or1,300mg (6.2%vs8.4%;p?0.03).43Thus,there is no con-vincing evidence from randomized studies that have compared different doses of aspirin that higher doses are more effective in reducing the risk of serious vascular events.In fact,both this limited set of randomized comparisons and the indirect compari-sons reported in the overview of the Antithrombotic Trialists’(ATT)Collaboration(Table2)are compat-ible with the reverse(ie,blunting of the antithrom-botic effect at higher doses of aspirin,consistent with dose-dependent inhibition of PGI2).Such inhibition of PGI2may be a potential mechanism by which COX-2inhibitors produce an excess risk of MI(see below).

The antithrombotic effects of a range of doses of aspirin also have been compared with an untreated control group in a number of thrombotic vascular disorders.The doses have varied between50and 1,500mg/d.Aspirin has been shown to be effective in the following conditions:unstable angina in which the incidence of acute MI or death was significantly reduced in four separate studies using daily doses of 75mg,32325mg,44650mg,45and1,300mg46;stable angina in which a dose of75mg/d reduced the incidence of acute MI or sudden death33;aortocoro-nary bypass surgery in which the incidence of early occlusion was similarly reduced with daily doses of 100mg,47325mg,48975mg,49and1,200mg49; thromboprophylaxis of patients with prosthetic heart valves who also received warfarin in whom the incidence of systemic embolism was reduced with daily doses of100mg,50500mg,51and1,500mg52,53; thromboprophylaxis of patients with arterial venous shunts undergoing long-term hemodialysis in whom a dose of160mg/d was shown to be effective54;acute MI in which a dose of162.5mg/d reduced early (35-day)mortality as well as nonfatal reinfarction and stroke55;transient cerebral ischemia in which doses between50and1,200mg/d were effective34,36,41,56–58; acute ischemic stroke in which doses of160to300 mg/d were effective in reducing early mortality and stroke recurrence59,60;and polycythemia vera in which100mg,37but not900mg,61was effective in reducing fatal and nonfatal vascular events. Thus,aspirin is an effective antithrombotic agent in doses between50and1,500mg/d.It is also possible from the results of the Dutch TIA study that 30mg/d is effective.42There is no evidence that low doses(50to100mg/d)are less effective than high doses(650to1,500mg/d)and,in fact,the opposite may be true.These clinical findings are consistent with saturability of platelet COX-1inactivation at doses as low as30mg/d.62

There is evidence,however,that doses of approx-imately300mg/d produce fewer GI side effects than doses of approximately1,200mg/d.41There is also some evidence that a dose of30mg/d produces fewer side effects than300mg/d.42The Clopidogrel in Unstable Angina To Prevent Recurrent Events (CURE)investigators have retrospectively investi-gated the relationship between the aspirin dose(the CURE protocol recommended75to325mg/d)and risk of major bleeding.63This study was a random-ized comparison of clopidogrel with placebo on a “background”of aspirin therapy.Patients with acute coronary syndromes receiving aspirin,?100mg/d, had the lowest rate of major or life-threatening bleeding complications both in the placebo(1.9%) and in the clopidogrel(3%)arms of the trial.Bleed-ing risks increased with increasing aspirin dose with or without clopidogrel.63

In summary,the saturability of the antiplatelet effect of aspirin at low doses,the lack of dose-response relationship in clinical studies evaluating its antithrombotic effects,and the dose dependence of its side effects all support the use of as low a dose of aspirin as has been found to be effective in the treatment of various thromboembolic disorders(Ta-ble1).Use of the lowest effective dose of aspirin(50 to100mg/d for long-term treatment)is currently the most appropriate strategy to maximize its efficacy and minimize its toxicity.6

2.3.2Effects of Aspirin Not Related to TXA2: Aspirin has been reported to have effects on hemostasis that are unrelated to its ability to inactivate platelet COX-1.These include dose-dependent inhibition of platelet function,64–68enhancement of fibrinolysis,69–71 and suppression of plasma coagulation.72–75

In contrast to the saturable and well-characterized (nanomolar aspirin concentration,rapid time course, physiologic conditions,single serine modification)

Table2—Indirect Comparison of Aspirin Doses Reducing Vascular Events in High-Risk

Patients(Section2.3.1)*

Aspirin Dose,

mg/d No.of

Trials

No.of

Patients

Odds

Reduction

500–1,5003422,45119?3%

160–3251926,51326?3%

75–150126,77632?6%

?7533,65513?8%

*Data are from Lindemann et al5/2001.

https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT203S

inhibition of COX-1by aspirin,13,62,76the putative mechanisms underpinning the non-PG effects of aspirin on hemostasis are dose dependent and less clearly defined.For example,inhibition of shear-induced platelet aggregation depends on the level of aspirin provided,and enhanced fibrinolysis due to N-acetylation of lysyl residues of fibrinogen is seen in vivo with high doses of aspirin(650mg bid)69and proceeds more rapidly in vitro under nonphysiologic alkaline conditions.77Aspirin suppresses plasma co-agulation through several mechanisms.The first, initially described in1943by Link et al and con-firmed by others,72,73is caused by an antivitamin K effect of aspirin.It requires very high doses of aspirin and does not contribute to the antithrombotic effect of aspirin when the drug is used in doses up to1,500 mg/d.The second is platelet dependent and is characterized by inhibition of thrombin generation in a whole blood system.74,75A single dose of500mg depresses the rate of thrombin generation,whereas repeated daily dosing with300mg of aspirin reduces the total amount of thrombin formed.78An interac-tion with platelet phospholipids,which is blunted in hypercholesterolemia,has been proposed to explain the effects of aspirin on thrombin generation.78It is possible(but unproven)that this effect occurs as a consequence of impaired platelet coagulant activity secondary to inhibition of TX-dependent platelet aggregation.It is unknown whether lower doses of aspirin are able to produce this effect.This sort of in vitro effect has been shown for other platelet inhibi-tors,such as glycoprotein(GP)-IIb/IIIa antagonists(see below).Furthermore,high-dose aspirin can cause ab-normal coagulation in vitro by direct acetylation of one or more clotting factors.This can be demonstrated in platelet-poor plasma and,thus,is not related to platelet inhibition or vitamin K antagonism.

Additional studies in both animal models and human subjects have reported antithrombotic effects of aspirin that may occur,at least in part,through mechanisms unrelated to inactivation of platelet COX-1.In animal models,Buchanan et al66and Hanson et al64reported that optimal antithrombotic activity of aspirin required doses in excess of those required to inhibit TXA2.In clinical studies,the results of a subgroup analysis of the North American Symptomatic Carotid Endarterectomy Trial study79 suggested that aspirin in doses of?650mg/d might be more effective than?325mg/d for the preven-tion of perioperative stroke in patients having carotid artery surgery.80This retrospective observation was refuted by a second prospective study,the Acetylsal-icylic Acid and Carotid Endarterectomy Trial,43 which tested the hypothesis that the wide area of collagen exposed by endarterectomy is a sufficiently strong stimulus to platelet aggregation to require a larger dose of aspirin.Approximately3,000patients scheduled for carotid endarterectomy were ran-domly assigned81,325,650,or1,300mg/d aspirin, started before surgery and continued for3months. The combined rate of stroke,MI,or death at3 months was significantly(p?0.03)lower in the low-dose groups(6.2%)than in the high-dose groups (8.4%)[primary analysis].There were no significant differences between the81-mg and325-mg groups or between the650-mg and1,300-mg groups in any of the secondary analyses of the data.43

A subgroup analysis of the Physicians’Health Study,81based on post hoc measurements of baseline plasma C-reactive protein performed in543appar-ently healthy men who subsequently had MI,stroke, or venous thrombosis,and in543study participants who did not report vascular complications,has found that the reduction in the risk of a first MI associ-ated with the use of aspirin(325mg on alternate days)appears to be directly related to the level of C-reactive protein,raising the possibility of antiin-flammatory as well as antiplatelet effects of the drug in cardiovascular prophylaxis.82This hypothesis is unlikely to be correct because,as noted above,the antiinflammatory effects of aspirin and other nonste-roidal antiinflammatory drugs(NSAIDs)are largely related to their capacity to inhibit COX-2activity induced in response to inflammatory cytokines,12as these clinical effects can be fully reproduced by highly selective COX-2inhibitors(coxibs)in patients with rheumatoid arthritis.83As shown in Table3,the dose and time dependence of the effects of aspirin on nucleated inflammatory cells expressing COX-2 vs anucleated platelets expressing COX-1are mark-edly different,thus making a clinically relevant anti-inflammatory effect of the drug at325mg every

Table3—Dose and Time Dependence of the Effects of Aspirin on Platelets and Inflammatory Cells(Section2.3.2)

Cellular Target Enzyme

Single

Dose,*mg

Duration of Prostanoid

Suppression,h

Cumulative Effects Upon

Repeated Dosing

Daily

Dose,?mg

Platelets COX-110024–48Yes50–81 Inflammatory cells COX-2?6503–4No3,000–5,000 *Dose causing full suppression of prostanoid formation and/or clinically detectable functional effect after single dosing.

?Range of doses shown clinically effective in long-term trials of cardiovascular protection or rheumatoid arthritis.

204S Antithrombotic and Thrombolytic Therapy8th Ed:ACCP Guidelines

other day pharmacologically implausible.Finally, aspirin has been reported to modify the way in which neutrophils and platelets84or erythrocytes and plate-lets85,86interact,to protect endothelial cells from oxidative stress,87and to improve endothelial dys-function in atherosclerotic patients.88However,nei-ther the molecular mechanism(s)nor the dose de-pendence of these effects have been clearly established.Although improved endothelial dysfunc-tion could reflect an antiinflammatory effect of aspirin of relevance to atherogenesis,it should be emphasized that the hypothesis has never been tested by an appropriately sized controlled prospec-tive study.

All of the evidence detailed above suggesting dose-dependent effects for aspirin is indirect and inconsistent with the failure to show a dose effect in randomized clinical trials and in the ATT overview analysis.7This failure to show a dose effect is the critical point of the discussion because it correlates with the saturability of the aspirin effect on platelet COX-1.For example,in studies with purified en-zyme and with isolated platelets,nanomolar concen-trations of aspirin will completely block PG synthesis within20min after exposure.89Higher concentra-tions and longer exposures will not alter the inhibi-tory effect of aspirin on PG synthesis because of this saturable quality.Exactly the same feature(maximal effect at low doses,absence of dose effect)is seen in clinical trials with aspirin as an antithrombotic agent. When one raises the dose of aspirin in this situation, no further or additional effect can be appreciated because the critical event has already taken place, namely,maximal inhibition of platelet TX synthesis. Thus,the consistency of dose requirements and saturability of the effects of aspirin in acetylating the platelet enzyme,8inhibiting TXA2production,25,62 and preventing atherothrombotic complications6,7 constitutes the best evidence that aspirin prevents thrombosis through inhibition of TXA2production. It is likely,therefore,that any of the potential effects of aspirin on other determinants of arterial throm-bosis are much less important than the inhibition of platelet COX-1activity.

2.3.3Aspirin“Resistance”:The term aspirin resis-tance has been used to describe a number of differ-ent phenomena,including the inability of aspirin to (1)protect individuals from thrombotic complica-tions,(2)cause a prolongation of the bleeding time, (3)reduce TXA2production,or(4)produce a typical effect on one or more in vitro tests of platelet function.90From a therapeutic standpoint,it is im-portant to establish whether aspirin resistance can be overcome by increasing the dose of aspirin,but unfortunately very few data bear directly on this issue.The fact that some patients may experience recurrent vascular events despite long-term aspirin therapy should be properly labeled as treatment failure rather than aspirin resistance.Treatment failure is a common phenomenon occurring with all drugs(eg,lipid-lowering or antihypertensive drugs). Given the multifactorial nature of atherothrombosis and the possibility that platelet-mediated thrombosis may not be responsible for all vascular events,it is not surprising that only a fraction(usually one fourth to one third)of all vascular complications can be prevented by any single preventive strategy.

It has been reported that a variable proportion(up to one fourth)of patients with cerebrovascular dis-ease only achieve partial inhibition of platelet aggre-gation at initial testing,and some(up to one third) seem to develop resistance to aspirin over time,even with increasing doses.91–93The results of these long-term studies carried out by Helgason et al are at variance with those of a short-term study of Weksler et al,94showing that aspirin,40mg/d,inhibited platelet aggregation and TXA2formation as effec-tively as higher doses of aspirin in patients who had recent cerebral ischemia.Variable platelet responses to aspirin have also been described in patients with peripheral arterial disease95and with ischemic heart disease.96–98In the Buchanan and Brister study,96 aspirin nonresponders were identified on the basis of bleeding time measurements.Approximately40%of patients undergoing elective coronary artery bypass grafting showed no prolongation of bleeding time in response to aspirin.This finding was associated with increased platelet adhesion and12-HETE synthe-sis.96In contrast,repeated measurements of platelet aggregation performed over24months of placebo-controlled treatment by Berglund and Wallentin99 demonstrated that100patients with unstable coro-nary artery disease randomized to receive aspirin, 75mg/d,in the Research Group on Instability in Coronary Artery Disease in Southeast Sweden study32had consistently reduced platelet aggregation without attenuation during long-term treatment. Based on measurements of platelet aggregation in response to arachidonate and adenosine diphosphate (ADP),5%and24%of patients with stable cardio-vascular disease who were receiving aspirin(325 mg/d for?7days)were defined as resistant and semiresponders,respectively.97Using a variety of techniques,including conventional aggregometry, shear stress-induced activation,and the expression of platelet surface receptors,Sane et al98recently re-ported that57%of a group of88patients with documented heart failure who had been treated with aspirin,325mg/d,for?1month showed aspirin nonresponsiveness.Overall,the majority of these studies were characterized by the following major

https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT205S

limitations:(1)biochemical or witnessed verification of patient’s adherence to the prescribed therapy were absent;(2)there was a single measurement of any given test;(3)intrasubject and intersubject variability and stability of the assay over time were usually not reported;(4)the criteria to define the normal vs the aspirin-resistant range and the assay conditions differed among studies;(5)doses of aspi-rin were heterogeneous,ranging from75to1,300 mg;and(6)none of these studies were properly controlled.

Lack of biochemical assessment of compliance is a major issue for the majority of studies assessing platelet function in response to aspirin,and this aspect is crucial in studies investigating aspirin unresponsiveness.Interestingly,a recent study100 in190patients with a history of MI compared arachidonate-induced platelet aggregation in pa-tients while receiving their usual aspirin therapy, after7days of withdrawal,and24h after a single witnessed intake of aspirin of325mg.Although9% of the patients who declared having taken their usual therapy failed to show inhibition of platelet aggrega-tion,this percentage dropped to?1%(1patient of 190)after a witnessed dose.100Furthermore,this single patient admitted NSAID intake12h before testing.Similar results were reported in the study by Lev et al,101where after a witnessed dose of325mg of aspirin,the mean of arachidonic acid-induced

light transmission aggregometry became?20%(the established limit to define resistance)in formerly resistant patients.Other studies have reported up to 40%noncompliance with long-term aspirin use.102It is therefore clear that questionnaires cannot be a reliable parameter to assess the compliance to any given treatment,including aspirin,and that studies not relying on salicylate measurements or serum TXB2have a major,intrinsic bias,seriously hamper-ing the interpretation of results.Furthermore,the few studies directly comparing different functional assays failed to find any significant agreement be-tween tests,generating the disappointing conclusion that aspirin nonresponsiveness may be highly test specific.

Several relatively small studies(n?39to180)of stroke patients103–105have suggested that aspirin resistance may contribute to treatment failure(ie, recurrent ischemic events while on antiplatelet ther-apy)and that doses higher than500mg may be more effective than lower doses in limiting this phenome-non.The uncontrolled nature and small sample size of these studies make it difficult to interpret the results.As noted above,a much larger database failed to substantiate a dose-dependent effect of aspirin in stroke prevention,7an effect that one would theoreti-cally expect if aspirin resistance could be overcome at least in part by increasing the daily dose of the drug. Gum et al106reported that5%of326stable cardiovascular patients were aspirin resistant based on the results of platelet aggregation induced by ADP and arachidonic acid.The aspirin-resistant group had an increased risk of death,MI,or cere-brovascular accident during almost2years of follow-up. There were,however,relatively few events in this study,and the rationale for the particular definition of aspirin resistance is uncertain.

Among a wide range of patients with vascular disease in whom the annual rate of serious vascular events ranges from40to80per1,000,aspirin typically prevents at least10to20fatal and nonfatal vascular events for every1,000patients treated for1 year.6Thus,30to60vascular events are expected to occur for every1,000patients treated with low-dose aspirin for1year not because of resistance,but because of the multifactorial nature of atherothrom-bosis.Thus,we do not agree with the definition given by Wang et al,107whereby“in its broadest sense,resistance refers to the continued occurrence of ischemic events despite adequate antiplatelet therapy and compliance.”Indeed,as suggested by Hennekens et al,108“given the multiple pathways by which platelets may be activated,it is perhaps more surprising that a clinical benefit is detectable in randomized trials of cardiovascular disease than that treatment failures complicate aspirin therapy.”

At least three potential mechanisms may underlie the occurrence of aspirin-resistant TXA2biosynthe-sis.The transient expression of COX-2in newly formed platelets in clinical settings of enhanced platelet turnover4is a potentially important mecha-nism that deserves further investigation.Extraplate-let sources of TXA2(eg,monocyte/macrophage COX-2)may contribute to aspirin-insensitive TXA2 biosynthesis in acute coronary syndromes.109Fur-thermore,Catella-Lawson et al110reported that con-comitant administration of a traditional NSAID(eg, ibuprofen)may interfere with the irreversible inac-tivation of platelet COX-1by aspirin.This is due to competition for a common docking site within the COX channel(arginine-120),which aspirin binds to with weak affinity before irreversible acetylation of Serine-529.13This pharmacodynamic interaction also has been described between naproxen and aspirin111 but does not occur with rofecoxib,110celecoxib,112or diclofenac,110drugs endowed with variable COX-2 selectivity.83Thus,concomitant treatment with readily available over-the-counter NSAIDs may limit the cardioprotective effects of aspirin and contribute to aspirin resistance.Based on current analysis of available data,113–115the US Food and Drug Admin-istration(FDA)has issued a statement informing

206S Antithrombotic and Thrombolytic Therapy8th Ed:ACCP Guidelines

patients and health-care professionals that ibuprofen can interfere with the antiplatelet effect of low-dose aspirin(81mg/d),potentially rendering aspirin less effective when used for cardioprotection and stroke prevention(https://www.360docs.net/doc/b711003600.html,/cder/drug/infopage/ ibuprofen/default.htm).

The clinical relevance of aspirin-resistant TXA2 biosynthesis has been explored by Eikelboom et al,116who performed a nested case-control study of baseline urinary TX metabolite excretion in relation to the occurrence of major vascular events in aspirin-treated high-risk patients enrolled in the Heart Outcomes Prevention Evaluation trial.After adjustment for baseline differences,the odds for the composite outcome of MI,stroke,or cardio-vascular death increased with each increasing quartile of11-dehydro-TXB2excretion,with pa-tients in the upper quartile having a1.8-times higher risk than those in the lower quartile.One limitation in this study,however,was the inability to differentiate between variable compliance in taking aspirin as prescribed(or avoiding NSAIDs)and variable occurrence of aspirin-resistant sources of TXA2biosynthesis.

Thus,in summary,both the mechanism(s)and clinical relevance of aspirin resistance,as defined by platelet aggregation measurements,remain to be established.117Until its true nature and preva-lence vis-a`-vis other antiplatelet drugs are better defined,no test of platelet function is recom-mended to assess the antiplatelet effect of aspirin in the individual patient.118,119On the other hand, additional studies on the mechanisms and clinical relevance of aspirin-resistant TXA2biosynthesis are clearly warranted.

2.4The Antithrombotic Effect of Aspirin

2.4.1Prevention of Atherothrombosis in Different Clinical Settings:The efficacy and safety of aspirin are documented from analysis of approximately70 randomized clinical trials that included?115,000 patients at variable risk of thrombotic complications of atherosclerosis.A detailed analysis of individual trials is beyond the scope of this article.It is more appropriately dealt within specific clinical sections of this volume.

Aspirin has been tested in patients demonstrating the whole spectrum of atherosclerosis,from appar-ently healthy low-risk individuals to patients present-ing with an acute MI or an acute ischemic stroke; similarly,trials have extended for as short as a few weeks’duration or as long as10years.6,7Although aspirin has been shown consistently to be effective in preventing fatal and/or nonfatal vascular events in these trials,both the size of the proportional effects and the absolute benefits of antiplatelet therapy are somewhat heterogeneous in different clinical settings. In the Second International Study of Infarct Sur-vival,55a single tablet of aspirin(162.5mg)started within24h of the onset of symptoms of a suspected MI and continued daily for5weeks produced highly significant reductions in the risk of vascular mortality (by23%),nonfatal reinfarction(by49%),and non-fatal stroke(by46%).There was no increase in hemorrhagic stroke or GI bleeding in the aspirin-treated patients and only a small increase in minor bleeding.55Treatment of1,000patients with sus-pected acute MI with aspirin for5weeks will result in approximately40patients in whom a vascular event is prevented,7with a proportional odds reduc-tion of30%(see the“Acute ST-Segment Elevation Myocardial Infarction”chapter).

Two separate trials with a similar protocol,the International Stroke Trial59and the Chinese Acute Stroke Trial,60tested the efficacy and safety of early aspirin use in acute ischemic stroke.Approximately 40,000patients were randomized within48h of the onset of symptoms to2to4weeks of daily aspirin therapy(300mg and160mg,respectively)or pla-cebo.An overview of the results of both trials suggests an absolute benefit of9fewer deaths or nonfatal strokes per1,000patients in the first month of aspirin therapy.7The proportional odds reduction in fatal or nonfatal vascular events is only10%in this setting.Although the background risk of hemorrhagic stroke was threefold higher in the Chinese Acute Stroke Trial than in the International Stroke Trial,the absolute increase in this risk associated with early use of aspirin was similar in the two studies(excess2per1,000 patients).59,60The broad clinical implications of these findings are discussed in the“Antithrombotic and Thrombolytic Therapy for Ischemic Stroke”chapter.In terms of their research implications,these results are consistent with biochemical evidence of episodic plate-let activation during the first48h after the onset of symptoms of an acute ischemic stroke and with sup-pression of in vivo TXA2biosynthesis in patients receiv-ing low-dose aspirin in this setting.120

Long-term aspirin therapy confers conclusive net benefit on risk of subsequent MI,stroke,or vascular death among subjects with high risk of vascular complications.These include patients with chronic stable angina,33patients with prior MI,7patients with unstable angina,32,44–46and patients with TIA or minor stroke34,36,41,56–58as well as other high-risk categories.7The proportional effects of long-term aspirin therapy on vascular events in these different clinical settings are rather homogeneous,ranging between20%and25%odds reduction based on an overview of all randomized trials.7However,individ-ual trial data show substantial heterogeneity,ranging

https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT207S

from no statistically significant benefits in patients with peripheral vascular disease to approximately 50%risk reduction in patients with unstable angina.7 Although other factors may play a role,we interpret these findings as reflecting the variable importance of TXA2as a mechanism amplifying the hemostatic response to plaque destabilization in different clini-cal settings.In terms of absolute benefit,these protective effects of aspirin translate into avoidance of a major vascular event in50per1,000patients with unstable angina treated for6months and in36 per1,000patients with prior MI,stroke,or TIA treated for approximately30months.7

For patients with different manifestations of ischemic heart or brain disease,a widespread consensus exists in defining a rather narrow range of recommended daily doses(ie,75to160mg)for the prevention of MI,stroke,or vascular death. This is supported by separate trial data in patients randomized to treatment with low-dose aspirin or placebo as well as by an overview of all antiplatelet trials showing no obvious dose dependence,from indirect comparisons,for the protective effects of aspirin7(Table3).There is no convincing evidence that the dose requirement for the antithrombotic effect of aspirin varies in different clinical settings. Among most high-risk patient groups,the ex-pected number avoiding a serious vascular event by using aspirin substantially exceeds the number expe-riencing a major bleed.However,it is unclear whether aspirin might benefit people who,although apparently healthy,are at intermediate risk of serious vascular events.The question of whether aspirin is effective for the primary prevention of vascular events has been addressed in a metaanalysis of randomized trials.332

Six primary prevention trials81,121–125including 92,873participants were studied(Table4).Mean follow-up was approximately6years.There was a 15%reduction in the odds of cardiovascular events (OR,0.85;95%CI,0.79to0.92;p?0.001)and highly significant reductions of23%in total coronary heart disease(CHD)[OR,0.77;95%CI,0.70to 0.86;p?0.001]and24%in nonfatal MI(OR,0.76; 95%CI,0.67to0.85;p?0.001).

There was no overall effect on stroke(OR,0.95; 95%CI,0.84to1.06;p?0.3),but data were not available separately for hemorrhagic and nonhemor-rhagic stroke,126–128so the effects on these two stroke subtypes could not be examined in detail. Aspirin had no significant effect on the aggregate of all vascular causes of death(OR,0.89;95%CI,0.72 to1.10;p?0.3),or on overall mortality(OR,0.94; 95%CI,0.87to 1.00;p?0.07).In summary, therefore,in primary prevention,aspirin chiefly pre-vents nonfatal MI,and appears to have little effect on fatal vascular events.

2.5Balance of Benefit and Harm

Previous metaanalyses of the effects of antiplatelet therapy among people at high risk of occlusive vascular disease7have shown that the benefits of aspirin far exceed the bleeding risks among such

Table4—Primary Prevention Trials(Section2.4.1)

Trial Dates of Recruitment

Year of

Publication

Mean

Duration of

Follow-up,*yr Target Population

Eligible

Age

Range,yr Intervention

Randomized

Factorial

Comparison

Placebo

Control

British

Doctors

Study

11/1/1978to11/1/19791988 5.6Male physicians19–90500mg/d None No

US physicians8/24/1981to4/2/19841988 5.0Male physicians45–73325mg

alternate

days Beta-carotene

(alternate days)

vs placebo

Yes

Thrombosis

Prevention

Trial 2/6/1989to5/18/19941998 6.7Men with risk

factors for

CHD

45–6975mg/d Warfarin vs placebo Yes

Hypertension Optimal

Treatment 10/12/1992to5/7/19941998 3.8Men and women

with diastolic

BP100–115

mm Hg

31–7575mg/d Three target diastolic

BPs(?80mm Hg,

?85mm Hg,

?90mm Hg)

Yes

Primary

Prevention

Project 6/8/1993to4/21/19982001 3.7Men and women

with one or

more risk

factors for

CHD

45–94100mg/d Vitamin E vs open

control

No

Women’s

Health

Study 9/1992to5/1995200510.1Female health

professionals

?45100mg

alternate

days

Vitamin E vs

placebo

Yes

*Mean duration of follow-up among surviving participants within each trial.

208S Antithrombotic and Thrombolytic Therapy8th Ed:ACCP Guidelines

patients.By contrast,the majority of participants (92%)in the primary prevention trials were at low absolute risk of coronary disease;on average,the annual risk of a vascular event in the primary pre-vention trials was only about one tenth of that occurring in the high-risk trials.Hence,although the proportional benefits of aspirin appeared broadly similar in primary and secondary prevention,the absolute benefits and risks of aspirin in the primary prevention trials were very small.Each year,fewer than1person in every1,000could expect to avoid an occlusive vascular event by taking aspirin,whereas a comparably small number could expect to experience a major extracranial bleed.The relative size of these opposing effects is too imprecisely known in low-risk people to predict the net public health consequences of widespread aspirin use in healthy people.The ATT Collaboration is currently analyzing individual partici-pant data from the six primary prevention trials,and these new analyses will help to clarify the benefits and risks of aspirin in particular groups of individuals.Until

the benefits of aspirin can be defined more precisely, however,the possibility of a benefit does not seem to justify the probability of a hazard.This emphasizes the need for trials of aspirin for primary prevention among specific groups at increased risk of vascular disease, such as people?70years of age and people with diabetes but no vascular disease.

2.5.1Atrial Fibrillation:Moderate-dose warfarin alone(international normalized ratio[INR],2.0to

3.0)is very effective in reducing the risk of stroke in patients with nonvalvular atrial fibrillation.129–131 The effectiveness of aspirin in doses between75and 325mg has been compared with warfarin and pla-cebo in three randomized trials of patients with non-valvular atrial fibrillation.130,132,133In one study,130 aspirin was significantly more effective than placebo, whereas in the other two132,133there was a nonsig-nificant trend in favor of aspirin.Pooled analysis of the three studies shows a relative risk reduction in favor of aspirin over placebo of about25%(range,14 to44%).Aspirin was significantly less effective than warfarin in two studies on an intention-to-treat anal-ysis,132,133and in the third study on an efficacy analysis.130On pooled analysis,warfarin was signifi-cantly more effective than aspirin,with a47%rela-tive risk reduction(range,28to61%;p?0.01).134 Moreover,adjusted-dose warfarin therapy(INR,2.0 to3.0)was more effective than fixed low-dose war-farin therapy(INR,1.2to1.5)and aspirin,325mg/d, in high-risk patients with atrial fibrillation.135Thus, aspirin appears to be effective in preventing stroke in patients with atrial fibrillation but is substantially less effective than warfarin.136,137

2.5.2Deep Vein Thrombosis:The Pulmonary Em-bolism Prevention Trial138has established that aspi-rin is effective in preventing venous thromboembo-lism after surgery for hip fracture.This was a double-blind multicenter study of13,356patients undergoing surgery for hip fracture and of an addi-tional4,088patients undergoing elective hip or knee arthroplasty.Patients were assigned160mg of aspi-rin or placebo qd for5weeks,with the first dose starting before surgery.Other forms of prophylaxis were allowed,and either heparin or low-molecular-weight heparin was used in about40%of the patients.Among the13,356patients with hip frac-ture,aspirin produced a36%reduction in symptom-atic deep vein thrombosis or pulmonary embolism (PE)[absolute risk reduction0.9%;p?0.0003].A similar relative risk reduction in patients who were assigned aspirin was observed in patients who also received heparin.

This important study,138therefore,clearly shows that aspirin reduces the incidence of fatal PE and symptomatic nonfatal deep vein thrombosis or PE in patients with hip fracture.The results of the Pulmo-nary Embolism Prevention trial are consistent with the meta-analysis performed by the Antiplatelet Tri-alists’Collaboration,139and supersede the findings in most of the previous trials.140–142However,in three randomized studies in major orthopedic surgery comparing aspirin with either warfarin143or a low-molecular-weight heparin,144,145the incidence of ve-nous thrombosis was significantly higher in the aspi-rin group in all three.

2.5.3Placental Insufficiency:The pathogenesis of preeclampsia and fetal growth retardation is related to reduced placental blood flow,which is believed to be caused by constriction and/or thrombosis of small placental arteries.146The initial reports that low-dose aspirin therapy reduces the risk of severe low birth weight among newborns,147and the risk of cesarean section in mothers with pregnancy-induced hyper-tension,146led to the widespread use of prophylactic aspirin to prevent preeclampsia.Subsequently,sev-eral larger trials reported no beneficial effects of aspirin.148–154

A systematic review155of data from39trials in ?30,000women showed that antiplatelet therapy (mostly aspirin,60mg/d)is associated with a15% decrease in the risk of preeclampsia.This effect was consistent,regardless of risk status(moderate or high),dose of aspirin,or gestation at trial entry. There was some evidence that there may be greater benefits for women given?75mg of aspirin,al-though the numbers of women in the subgroup were small and so a potential for random error.There was also an8%reduction in the risk of preterm birth and

https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT209S

a14%reduction in the risk of fetal or neonatal death for women allocated antiplatelet therapy.155Remain-ing questions are whether particular subgroups of high-risk women might have greater benefit and whether earlier treatment(ie,before12weeks)or aspirin doses of?75mg would have additional benefits without an increase in adverse effects.155 The potential involvement of extra platelet sources of vasoactive eicosanoids expressing COX-2in response to a local growth-promoting milieu might contribute, at least in part,to the limited efficacy of low-dose aspirin therapy in this setting.

2.6Adverse Effects of Aspirin

Aspirin does not cause a generalized bleeding abnormality unless it is given to patients with an underlying hemostatic defect,such as hemophilia, uremia,or that induced by anticoagulant therapy. Aspirin-induced impairment of primary hemostasis cannot be separated from its antithrombotic effect and is similar at all doses?75mg/d.6

The balance between preventing vascular occlu-sion and causing excess bleeding with aspirin de-pends critically on the absolute thrombotic vs hem-orrhagic risk of the patient.Thus,in individuals at low risk for vascular occlusion(eg,?1%/yr),a very small absolute benefit is offset by exposure of a large number of healthy subjects to undue bleeding com-plications.In contrast,in patients at high risk of cardiovascular or cerebrovascular complications(eg,?3%/yr),the substantial absolute benefit of aspirin prophylaxis clearly outweighs the harm(Table5). For example,the absolute excess of major bleeds(ie, those requiring transfusion)in acute MI is approxi-mately1/100th the absolute number of major vascu-lar events avoided by aspirin therapy.7

The overall risk of major extracranial and intracra-nial hemorrhage associated with antiplatelet drugs is difficult to assess in individual trials because their incidence is low(ie,?1%/yr),making detection of even a50to60%relative increase in risk unrealistic in most trials of a few thousand patients.

Aspirin-induced GI toxicity,as detected in ran-domized clinical trials,appears to be dose related in the range of30to1,300mg/d.156This,along with studies of the relationship of efficacy to dose,is based largely on indirect comparisons of different trials and on a limited number of randomized,direct comparisons of different aspirin doses,as reviewed above.Such a dose-response relationship is thought to reflect at least two COX-1-dependent compo-nents,dose-dependent inhibition of COX-1in the GI mucosa and dose-independent(within the range of examined doses)inhibition of COX-1in platelets.6 Thus,it is not surprising that the antithrombotic effect of aspirin can be dissociated,at least in part, from its most common side effect.However,even when administered at low doses,aspirin can cause serious GI bleeding,as reported in studies using30 to50mg/d.36,42Because of the underlying preva-lence of gastric mucosal erosions related to concur-rent use of other NSAIDs and/or Helicobacter pylori infection in the general population,it should be expected that any antiplatelet dose of aspirin will cause more bleeding from preexisting lesions than a placebo.Consistent with this mechanistic interpre-tation,the relative risk of hospitalization due to upper-GI bleeding and/or perforation associated with low-dose aspirin therapy(mostly,100to300 mg/d)is comparable to that of other antiplatelet agents and anticoagulants(ie,2.3[95%CI,1.7to 3.2],2.0[95%CI,1.4to2.7],and2.2[95%CI,1.4to 3.4],respectively)in a large population-based obser-vational study.157

In the overview of the ATT Collaboration,7infor-mation was available on787major extracranial hem-orrhages in60trials recording at least one such hemorrhage.These were generally defined as hem-orrhages that were fatal or required transfusion; among them,159(20%)caused death.Overall,the proportional increase in risk of a major extracranial bleed with antiplatelet therapy was about one half (odds ratio[OR],1.6;95%CI,1.4to1.8),with no significant difference between the proportional in-creases observed in each of the five high-risk cate-gories of patients.After allowing for noncompliance in the trials,they are compatible with the2-to

2.5-fold excess observed in case-control studies.

A case-control study with hospital and community controls has examined the risks of hospitalization for

Table5—Benefit and Harm of Antiplatelet Prophylaxis

With Aspirin in Different Settings

(Section2.6)

Clinical Setting Benefits*Harm?

Men at low to high cardiovascular risk1–21–2

Essential hypertension1–21–2

Chronic stable angina101–2

Prior MI201–2

Unstable angina501–2

*No.of patients in whom a major vascular event is avoided per

1,000/yr.Benefits are calculated from randomized trial data re-

viewed in this article and depicted in Figure3.

?No.of patients in whom a major GI bleeding event is caused per

1,000/yr.Excess of upper-GI bleedings is estimated from a back-

ground rate of1event per1,000/yr in the general population of

nonusers157and an RR of 2.0to 3.0associated with aspirin

prophylaxis.157–159Such an estimate assumes comparability of other

risk factors for upper-GI bleeding,such as age and concomitant use

of NSAIDs,and may actually underestimate the absolute risk in an

elderly population exposed to“primary”prevention.

210S Antithrombotic and Thrombolytic Therapy8th Ed:ACCP Guidelines

bleeding peptic ulcer associated with three different regimens of aspirin prophylaxis.158ORs were raised for all doses of aspirin taken:75mg,OR2.3(95%CI, 1.2to4.4);150mg,OR3.2(95%CI,1.7to6.5); and300mg,OR3.9(95%CI,2.5to6.3).Addi-tional epidemiologic studies have found a dose-response relationship between aspirin prescription and upper-GI complications,as reviewed by Garc?′a Rodr?′guez et al.159It has been calculated that ap-proximately900of the10,000episodes of ulcer bleeding occurring in people?60years of age each year in England and Wales could be associated with, and ascribed to,prophylactic aspirin use.158A gen-eral change to lower doses of aspirin(75mg)would not eliminate risks but would reduce risk by about 40%compared with300-mg doses and by30% compared with150-mg doses if the assumptions from indirect comparisons are correct.158Given that the mortality rate among patients who are hospital-ized for NSAID-induced upper-GI bleeding is about 5to10%,160,161such a strategy could save a signifi-cant number of lives.

The widely held belief that enteric-coated and buffered varieties of aspirin are less likely to occasion major upper-GI bleeding than plain tablets was tested in data from a multicenter case-control study.162The relative risks of upper-GI bleeding for plain,enteric-coated,and buffered aspirin at average daily doses of?325mg were2.6,2.7,and3.1, respectively.At doses?325mg,the relative risks were5.8for plain and7.0for buffered aspirin;there were insufficient data to evaluate enteric-coated aspirin at this dose level.162Similar conclusions were reached by a case-control study using data from the UK General Practice Research Database.163Thus, physicians who recommend aspirin in an enteric-coated or buffered form should not assume that these formulations are less likely to cause GI tract bleeding than plain aspirin.

Suppressing acid secretion is thought to reduce the risk of ulcers associated with regular use of NSAIDs.In patients who required continuous treat-ment with NSAIDs and who had ulcers or?10 erosions in either the stomach or duodenum,ome-prazole healed and prevented ulcers more effectively than did ranitidine.164In these patients,maintenance therapy with omeprazole was associated with a lower rate of relapse and was better tolerated than miso-prostol.165In high-risk patients(history of previous ulcer bleeding)taking low-dose aspirin for6months, omeprazole and H pylori eradication were associated with similar rates of recurrent bleeding(0.9%vs 1.9%),166although clinically important differences between the two preventive strategies could not be excluded owing to the small sample size(n?250). Two relatively small studies167,168have challenged current guidelines that recommend clopidogrel for patients who have major GI contraindications to aspirin,principally recent significant bleeding from a peptic ulcer or gastritis.169,170Both studies enrolled patients with ulcer bleeding after the use of low-dose aspirin.In the study of Chan et al,167after healing of ulcers and eradication of H pylori,if present,320 patients were randomly assigned to receive either clopidogrel,75mg/d,or aspirin,80mg/d,plus20mg bid of esomeprazole for12months.The cumulative incidence of recurrent bleeding was8.6%(95%CI, 4.1to13.1%)among patients who received clopi-dogrel and0.7%(95%CI,0to2.0%)among those who received aspirin plus esomeprazole(p?0.001).167 In the study of Lai et al,168170patients with prior ulcer bleeding were randomly assigned to treatment with clopidogrel,75mg/d,or aspirin,100mg/d,and esome-prazole,20mg/d,for1year.The cumulative incidence of recurrent ulcer complications was13.6%and0%, respectively(95%CI for the difference,6.3to20.9%; p?0.0019).168The consistent findings of two indepen-dent studies suggest that the combination of esomepra-zole and low-dose aspirin is superior to clopidogrel in preventing recurrent ulcer bleeding in patients with a history of aspirin-related ulcer bleeding. Substantially less information is available about the risk of intracranial hemorrhage associated with aspirin use.In the Nurses’Health Study171cohort of approximately79,000women34to59years of age, infrequent use of aspirin(1to6tablets per week) was associated with reduced risk of ischemic stroke, whereas high frequency of use(?15aspirin tablets per week)was associated with increased risk of subarachnoid hemorrhage,particularly among older or hypertensive women.In the overview of the ATT Collaboration,7the overall absolute excess of intra-cranial hemorrhage due to aspirin therapy was?1 per1,000patients per year in high-risk trials,with somewhat higher risks in patients with cerebrovas-cular disease.

Low-dose aspirin therapy has not been reported to affect renal function or BP control,172consistent with its lack of effect on renal prostaglandins173that derive primarily from constitutively expressed COX-2in the human kidney.83Moreover,aspirin,75mg/d,did not affect BP or the need for antihypertensive therapy in intensively treated hypertensive patients.122The sug-gestion that the use of aspirin and other antiplatelet agents is associated with reduced benefit from ena-lapril in patients with left ventricular systolic dys-function174is not supported by the results of a large metaanalysis of MI trials.175Similarly,no negative interaction occurs between angiotensin-converting enzyme(ACE)inhibition and the cardiovascular benefits of low-dose aspirin in intensively treated hypertensive patients.176The ACE Inhibitors Col-

https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT211S

laborative Group 177has performed a systematic overview of data for 22,060patients from six long-term randomized trials of ACE inhibitors to assess whether aspirin altered the effects of ACE inhibitor therapy on major clinical outcomes.Even though results from these analyses cannot rule out the possibility of some sort of interaction,they show unequivocally that even if aspirin is given,the addi-tion of ACE inhibitor therapy produced substantial additional benefit in all major vascular outcomes.Therefore,in the absence of clear contraindications,concomitant use of aspirin and ACE inhibitors should be considered in all patients at high risk of major vascular events.177

Thus,in summary,inhibition of TXA 2-dependent platelet function by aspirin is effective for the pre-vention of thrombosis,but is also associated with excess bleeding.Assessing the net effect requires an estimation of the absolute thrombotic vs hemor-rhagic risk of the individual patient.In individuals at very low risk for vascular occlusion,a very small absolute benefit may be offset by exposure of very large numbers of healthy subjects to undue bleeding complications.As the risk of experiencing a major vascular event increases,so does the absolute benefit of antiplatelet prophylaxis with aspirin,as shown in Figure 3,for a number of clinical settings in which the efficacy of the drug has been tested in random-ized clinical trials.Based on the results of such trials,

the antithrombotic effect of aspirin does not appear to be dose related over a wide range of daily doses (30to 1,300mg),an observation consistent with saturability of platelet COX inhibition at very low doses.In contrast,GI toxicity of the drug does appear to be dose related,consistent with dose-and dosing interval-dependent inhibition of COX activity in the nucleated lining cells of the GI mucosa.Thus,aspirin once daily should be considered in all clinical conditions in which antiplatelet prophylaxis has a favorable benefit/risk profile.Because of GI toxicity and its potential impact on compliance,physicians are encouraged to use the lowest dose of aspirin shown effective in each clinical setting (Table 1).2.7Reversible COX Inhibitors

In the absence of definitive randomized studies,traditional NSAIDs have long been thought to pose no cardiovascular hazard or to be somewhat cardio-protective.Because of their reversible mechanism of action in inhibiting platelet COX-1and of their short half-lives,most traditional NSAIDs inhibit TXA 2-dependent platelet activation only transiently and incompletely in the vast majority of users.178A notable exception is provided by naproxen,which when administered regularly at 500mg bid,has been shown to inhibit TXA 2biosynthesis in vivo to the same extent as low-dose aspirin,179consistent with its

Annual risk of a vascular event on placebo

010********

600

5101520

Subjects in whom a vascular event is prevented by

aspirin

per 1,000 treated

/yr

Figure 3.The absolute risk of vascular complications is the major determinant of the absolute benefit of antiplatelet prophylaxis.Data are plotted from placebo-controlled aspirin trials in different clinical settings.For each category of patients,the abscissa denotes the absolute risk of experiencing a major vascular event as recorded in the placebo arm of the trial(s).The absolute benefit of antiplatelet treatment is reported on the ordinate as the number of subjects in whom an important vascular event (nonfatal MI,nonfatal stroke,or vascular death)is actually prevented by treating 1,000subjects with aspirin for 1year.

212S

Antithrombotic and Thrombolytic Therapy 8th Ed:ACCP Guidelines

relative COX-1selectivity and longer half-life than other commonly used NSAIDs.

The only reversible COX inhibitors that have been tested in randomized clinical trials for their antithrom-botic efficacy are sulfinpyrazone,indobufen,flurbipro-fen,and triflusal.Sulfinpyrazone is a uricosuric agent structurally related to the antiinflammatory agent phe-nylbutazone.When used at the highest approved dosage of200mg qid,the drug inhibits platelet COX activity by approximately60%,after conversion from an inactive sulfoxide to an active sulfide metabo-lite.180The conflicting or negative results obtained in randomized clinical trials of sulfinpyrazone in pa-tients with MI or unstable angina7(reviewed in the “Valvular and Structural Heart Disease”chapter)are not surprising in light of the drug being a weak COX inhibitor with no other established antiplatelet mechanism of action.

In contrast,indobufen is a very potent inhibitor of platelet COX-1activity and has comparable bio-chemical,functional,and clinical effects to those of a standard dose of aspirin.Thus,at therapeutic plasma levels achieved after oral dosing of200mg bid, indobufen inhibits serum TXB2by?95%through-out the dosing interval181and reduces urinary TX metabolite excretion to an extent quite comparable to aspirin.182The finding that indobufen is as effec-tive as aspirin in preventing coronary graft occlusion in two randomized trials183,184is mechanistically consistent with the concept of platelet COX-1inhi-bition largely accounting for the antithrombotic ef-fect of aspirin,as discussed above.Indobufen also has been investigated in a small placebo-controlled study of patients with heart disease at increased embolic risk185and compared with warfarin186and ticlopidine187in patients with nonrheumatic atrial fibrillation and patients with recent reversible cere-bral ischemia,respectively.However,none of these studies in?4,000patients clearly established an advantage of indobufen vs standard treatments,al-though the95%CIs for these comparisons are wide. Indobufen has been reported to suppress in vivo TXA2biosynthesis more effectively than low-dose aspirin in patients with unstable angina,an effect possibly related to inhibition of monocyte COX-2by therapeutic plasma levels of indobufen.14The clini-cal relevance of these findings remains to be estab-lished.

Flurbiprofen has been evaluated in a single placebo-controlled,randomized trial of461patients with acute MI.188The6-month reinfarction rate was significantly lower in the flurbiprofen group(3%) than in the placebo group(10.5%),with an extremely low mortality rate(1.1%)in both groups.The small sample size of the study limits interpretation of these findings.

Triflusal,a salicylic acid derivative,reversibly in-hibits platelet COX activity after conversion to a long-lived metabolite,2-hydroxy-4-trifluoromethyl-benzoic acid.189Although the half-life of the parent

compound is only about30min,that of the deacety-lated metabolite approximates2days.Although tri-flusal is claimed to have negligible effects on vascular PGI2production,this is likely to reflect the exper-imental conditions used for the assessment of PGI2production ex vivo.The limited sample size of head-to-head comparisons of triflusal vs aspirin in patients randomized within24h of acute MI190 and in patients with cerebrovascular disease191 precludes unequivocal interpretation of the similar rates of major vascular events in the two treatment groups.None of these reversible COX inhibitors are approved as an antiplatelet drug in the United States,and it is unclear under which circum-stances they are prescribed instead of aspirin in other countries.

2.8Coxibs and Cardiovascular Disease

Coxibs were developed in an attempt to prevent the adverse GI effects of nonselective NSAIDs(by avoiding inhibition of COX-1)while maintaining equivalent antiinflammatory efficacy(by inhibiting COX-2).83Several large randomized trials192–194have demonstrated that coxibs are associated with lower risk of serious GI events than nonselective NSAIDs, but the Vioxx GI Outcomes Research Study193 among approximately8,000patients with rheuma-toid arthritis showed that those allocated to rofe-coxib,50mg/d,experienced a higher risk of vascular events than those allocated to naproxen500mg bid. This excess was almost entirely accounted for by a difference in the incidence of MI(20in2,699 person-years of follow-up among rofecoxib-allocated patients,vs4in2,699person-years among naproxen-allocated patients).There were no significant differ-ences in stroke(11rofecoxib vs9naproxen)or vascular deaths(7rofecoxib vs7naproxen).193Three placebo-controlled trials have now revealed a two-fold-to threefold-increased risk of vascular events in approximately6,000patients treated short term(10 days)with valdecoxib195or long term(up to3years) with celecoxib196or rofecoxib197both with and with-out concomitant aspirin treatment.These recent findings are consistent with a mechanism-based car-diovascular hazard for the class198and have led to the withdrawal of rofecoxib and valdecoxib from the market.

A metaanalysis of tabular data from138random-ized trials of five different coxibs in approximately 145,000patients has revealed that in placebo com-parisons,allocation to a coxib was associated with a

https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT213S

42%increased incidence of vascular events with no statistically significant heterogeneity among the dif-ferent coxibs.20This excess risk of vascular events was derived primarily from a twofold-increased risk of MI.Overall,there was no significant difference in the incidence of vascular events between a coxib and any traditional NSAID,but there was evidence of a significant difference between naproxen and the other traditional NSAIDs.20Given the nonlinear relationship between inhibition of platelet COX-1 activity and inhibition of platelet activation in vivo (Fig2),31it is perhaps not surprising that the cardio-vascular safety profile of coxibs and some non-naproxen NSAIDs(primarily diclofenac and ibupro-fen)appears similar because these drugs fail to inhibit platelet activation adequately irrespective of their COX-2selectivity.The results of the Multina-tional Etoricoxib and Diclofenac Arthritis Long-Term study,199comparing long-term treatment with etoricoxib and diclofenac in approximately35,000 arthritis patients,are consistent with this conclusion. Whether the variable level and duration of COX-1 inhibition by different NSAIDs modulate the cardio-vascular consequences of COX-2inhibition presently is unknown,given the limited utilization of NSAIDs other than ibuprofen,diclofenac,and naproxen in coxib trials.Thus,coxibs and some traditional NSAIDs moderately increase the risk of vascular events, particularly MI,but there remains considerable un-certainty about the magnitude of this hazard for particular drug regimens and patients subgroups.A metaanalysis of individual participant data from ran-domized coxib trials is currently being conducted by the Coxib Trialists’Collaboration in order to address some of the open questions related to the influence of dose,duration,and baseline characteristics,in-cluding the concomitant use of low-dose aspirin,on this cardiotoxicity.

3.0Dipyridamole Dipyridamole is a pyrimidopyrimidine derivative with vasodilator and antiplatelet properties.The mechanism of action of dipyridamole as an antiplate-let agent has been a subject of controversy.200Both inhibition of cyclic nucleotide phosphodiesterase (the enzyme that degrades cyclic adenosine mono-phosphate?AMP?to5(1)-AMP,resulting in the intraplatelet accumulation of cyclic AMP,a platelet inhibitor)and blockade of the uptake of adenosine (which acts at A2receptors for adenosine to stimu-late platelet adenylyl cyclase and thus increase cyclic AMP)have been suggested.Moreover,direct stim-ulation of PGI2synthesis and protection against its degradation have been reported,although the dipy-ridamole concentrations required to produce these effects far exceed the low micromolar plasma levels achieved after oral administration of conventional doses(100to400mg/d).200Dipyridamole also dif-ferentially inhibits the expression of critical inflam-matory genes in platelet-leukocyte aggregates.201 The absorption of dipyridamole from conventional formulations is quite variable and may result in low systemic bioavailability of the drug.A modified-release formulation of dipyridamole with improved bioavailability has been developed in association with low-dose aspirin.202Dipyridamole is eliminated pri-marily by biliary excretion as a glucuronide conjugate and is subject to enterohepatic recirculation.A ter-minal half-life of10h has been reported.This is consistent with the twice-daily regimen used in recent clinical studies.

Although the clinical efficacy of dipyridamole, alone or in combination with aspirin,has been questioned on the basis of earlier randomized tri-als,2,203the whole issue has been reopened by the reformulation of the drug to improve bioavailability and the results of the ESPS-2and European Stroke Prevention Reversible Ischemia Trial(ESPRIT) studies.36,204In ESPS-2,the new preparation of dipyridamole was evaluated in6,602patients with prior stroke or TIA.36This study showed that the addition of modified-release dipyridamole200mg bid to aspirin25mg bid was associated with a22% relative risk reduction of major vascular events com-pared with aspirin alone.Headache was the most common adverse effect of dipyridamole.Bleeding at any site was almost doubled in the two aspirin arms but was surprisingly indistinguishable from placebo in the dipyridamole-treated patients.36In a post hoc analysis of cardiac events in patients with CHD or MI at entry,dipyridamole did not result in a higher number of fatal and nonfatal cardiac events.204 More recently,the ESPRIT Study Group205has performed a randomized trial in which they assigned 2,739patients within6months of a TIA or minor stroke of presumed arterial origin to aspirin(30to 325mg/d)with or without dipyridamole(200mg bid).The primary outcome(a composite of major vascular events or major bleeding complications)was significantly reduced by the combined treatment vs aspirin alone by20%.Patients receiving aspirin and dipyridamole discontinued trial medication almost three times more often than those receiving aspirin alone,mainly because of headache.205Addition of the ESPRIT data to the metaanalysis of previous trials resulted in an overall risk ratio of0.82(95%CI, 0.74to0.91)for the composite of vascular death, stroke,or MI.However,based on the most recent Cochrane review,203the additional benefit of the combination over aspirin alone is not detectable in

214S Antithrombotic and Thrombolytic Therapy8th Ed:ACCP Guidelines

patients with other types of vascular disease. Whether this apparent discrepancy reflects a differ-ent prevalence of dipyridamole-sensitive mecha-nisms of disease or,perhaps more likely,the differ-ent types of formulation and daily dosage of the drug remains to be established.The fixed combination of modified-release dipyridamole and low-dose aspirin has been approved for stroke prevention by the FDA and other regulatory authorities.

4.0Thienopyridines Ticlopidine and clopidogrel are structurally re-lated thienopyridines with platelet-inhibitory prop-erties.Both drugs selectively inhibit ADP-induced platelet aggregation with no direct effects on arachi-donic acid metabolism.206Although ticlopidine and clopidogrel also can inhibit platelet aggregation in-duced by collagen and thrombin,these inhibitory effects are abolished by increasing the agonist con-centration and,therefore,are likely to reflect block-ade of ADP-mediated amplification of the platelet response to other agonists.

Neither ticlopidine nor clopidogrel affect ADP-induced platelet aggregation when added in vitro,up to500?mol/L,thus suggesting that in vivo hepatic transformation to an active metabolite(s)is necessary for their antiplatelet effects.In the liver,clopidogrel is metabolized into2-oxo-clopidogrel through a cy-tochrome P450-dependent pathway.This intermedi-ate metabolite is then hydrolyzed and generates the highly labile active metabolite,207which reacts as a thiol reagent with the ADP receptors on platelets when they pass through the liver.208The active metabolite belongs to a family of eight stereoiso-mers,only one of which(bearing7S,3Z,and4S or 4R configuration)retains biological activity.208 Experimental evidence suggests that clopidogrel and,probably,ticlopidine induce irreversible alter-ations of the platelet receptor P2Y12mediating inhibition of stimulated adenylyl cyclase activity by ADP.209,210The active metabolite of clopidogrel couples through a disulfide bridge to the P2Y12 receptor,presumably to the cysteine residue in the first extracellular loop;this results in oligomers dis-sociating into dimeric receptors that are partitioned out of lipid rafts,thereby losing the ability to bind their endogenous ligand.211Interestingly,mutations in the P2Y12gene are associated with a congenital bleeding disorder and abnormality in the platelet response to ADP,resembling that induced by thienopyridines.212Permanent modification of a platelet ADP receptor by thienopyridines is con-sistent with time-dependent cumulative inhibition of ADP-induced platelet aggregation on repeated daily dosing with ticlopidine or clopidogrel and with slow recovery of platelet function after drug withdrawal.206

4.1Ticlopidine

Up to90%of a single oral dose of ticlopidine is rapidly absorbed in humans.206Peak plasma concen-trations occur1to3h after a single oral dose of250 mg.Plasma levels of ticlopidine increase by approx-imately threefold on repeated twice-daily dosing over2to3weeks because of drug accumulation. Greater than98%of ticlopidine is reversibly bound to plasma proteins,primarily albumin.Ticlopidine is metabolized rapidly and extensively.A total of13 metabolites have been identified in humans.Of these,only the2-keto derivative of ticlopidine is more potent than the parent compound in inhibiting ADP-induced platelet aggregation.206

The apparent elimination half-life of ticlopidine is 24to36h after a single oral dose and up to96h after 14days of repeated dosing.206The standard regimen of ticlopidine is250mg bid,although it is unclear how a twice-daily regimen is related to the pharma-cokinetic and pharmacodynamic features noted above.A delayed antithrombotic effect was noted in at least one clinical trial of ticlopidine in patients with unstable angina with no apparent protection during the first2weeks of drug administration.213 Therefore,ticlopidine is not useful when a rapid antiplatelet effect is required.

Ticlopidine as a single agent has been evaluated in patients with stroke,214transient cerebral isch-emia,215unstable angina,213MI,216intermittent clau-dication,217–219and aortocoronary bypass surgery.220 Ticlopidine was significantly(but marginally in abso-lute terms)more effective than aspirin in reducing stroke in patients with transient cerebral ischemia or minor stroke215(although there was no statistically significant difference in the combined outcome of stroke,MI,or death)7;was as effective as aspirin in the treatment of patients with a recent MI216;was more effective than placebo in reducing the risk of the combined outcome of stroke,MI,or vascular death in patients with thromboembolic stroke214;was more effective than conventional antianginal therapy in reducing vascular death or MI in patients with unstable angina213;was more effective than placebo in reducing acute occlusion of coronary bypass grafts220;and was more effective than controls in improving walking distance218and reducing vascular complications in patients with peripheral vascular disease.217–219The association of ticlopidine therapy with hypercholesterolemia and neutropenia(for which the reported rate of occurrence is2.4%for neutrophils?1.2?109/L and0.8%for neutrophils

https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT215S

?0.45?109/L)and its comparative expense have reduced enthusiasm for this therapy as an alternative to aspirin in most situations.221Ticlopidine also has been associated with thrombocytopenia,221aplastic anemia,222and thrombotic thrombocytopenic pur-pura(TTP).223Ticlopidine has been approved for clinical use in patients with cerebral ischemia when aspirin has failed,cannot be tolerated,or is contra-indicated,although this limitation does not apply to all countries where the drug is registered. Additive effects of ticlopidine and aspirin have been described in rats,in inhibition of ADP-induced platelet aggregation ex vivo,tail bleeding time pro-longation,and protection from thrombosis in exper-imental models of platelet-dependent vascular occlu-sion.224Additive antiplatelet effects of aspirin(40 mg)and ticlopidine(250mg)have been reported in healthy volunteers.225Two studies226,227have dem-onstrated the superiority of ticlopidine with aspirin compared to aspirin alone or aspirin plus warfarin in preventing thrombotic complications after coronary artery stent placement.Ticlopidine has been rou-tinely used in combination with aspirin in patients receiving coronary artery stents,but the better safety profile of clopidogrel has resulted in the replacement of clopidogrel for ticlopidine as the standard anti-platelet regimen after stent deployment.228The risk of TTP associated with ticlopidine use has been estimated as0.02%in patients receiving the drug after stent placement.229This risk compares with an incidence of0.0004%in the general population.The mortality rate for this rare complication exceeds 20%.229The place of ticlopidine in the current therapeutic armamentarium is uncertain:(1)the drug is not uniformly cheaper than clopidogrel in different countries;(2)in contrast to clopidogrel, ticlopidine has no approved indication for the long-term management of post-MI patients;(3)ticlopi-dine has a higher bone-marrow toxicity than clopi-dogrel;and(4)because of safety concerns,an adequate loading dose of ticlopidine,as required in the acute setting,is unlikely to be used.

4.2Clopidogrel

The pharmacokinetics of clopidogrel are somewhat different from those of ticlopidine.Thus,after admin-istration of single oral doses(up to200mg)or repeated doses(up to100mg/d),unchanged clopidogrel was not detectable in peripheral venous plasma.230Concentra-tions of1to2ng/mL were measured in the plasma of patients who received150mg/d of clopidogrel (twice as much as the dose used in the Clopidogrel vs Aspirin in Patients at Risk of Ischemic Events [CAPRIE]study231and approved for clinical use)for 16days.The main systemic metabolite of clopidogrel is the carboxylic acid derivative,SR26334.Based on measurements of circulating levels of SR26334,it has been inferred that clopidogrel is rapidly absorbed and extensively metabolized.230The plasma elimination half-life of SR26334is approximately8h.As noted above,clopidogrel,inactive in vitro,is metabolically transformed by the liver into a short-lived active plate-let inhibitor.However,the interindividual variability in this metabolic activation is still being assessed,and to our knowledge,there are no published data on whether liver impairment decreases the ability of clopidogrel to inhibit platelet function.A pharmacokinetic/pharmacody-namic study232of a600-mg loading dose of clopi-dogrel in healthy subjects revealed linear correla-tions between the maximal antiplatelet effect and peak plasma concentrations of unchanged clopi-dogrel,of the carboxyl metabolite,and of the thiol metabolite as well as linear correlations between peak plasma concentration values of clopidogrel and its metabolites.These results have been interpreted to suggest that the pharmacodynamic response vari-ability is predominantly caused by individual differ-ences in clopidogrel absorption.232Because the cytochrome P450isozymes CYP3A4and3A5metab-olize clopidogrel faster than other human P450 isozymes and are the most abundant P450s in human liver,they are predicted to be predominantly respon-sible for the activation of clopidogrel in vivo.233 When clopidogrel and atorvastatin,a CYP3A4sub-strate,are present at equimolar concentrations in vitro,clopidogrel metabolism is inhibited by ?90%.233Variable metabolic activity of CYP3A4 may contribute to the interindividual variability in the platelet inhibitory effect of clopidogrel.234Thus, Angiolillo et al235recently characterized the influ-ence of CYP3A4genotype on interpatient variability in clopidogrel responsiveness.An intronic single nucleotide polymorphism in the CYP3A4gene, IVS10?12G?A(also called CYP3A4*1G)modified platelet reactivity ex vivo as measured by GP-IIb/IIIa receptor activation in response to clopidogrel in a group of patients with stable CHD receiving long-term anti-platelet therapy.The findings were replicated in a group of clopidogrel-na?¨ve patients undergoing elective percutaneous coronary intervention(PCI)treated with a300-mg loading dose of the drug.235 Clopidogrel inhibited ADP-induced platelet ag-gregation in a dose-dependent fashion with an ap-parent ceiling effect(40%inhibition)at400mg after single oral doses in healthy volunteers.Inhibition of platelet aggregation was detectable2h after oral dosing of400mg and remained relatively stable up to48h.230On repeated daily dosing of50to100mg of clopidogrel to healthy volunteers,ADP-induced platelet aggregation was inhibited from the second day of treatment(25to30%inhibition)and reached

216S Antithrombotic and Thrombolytic Therapy8th Ed:ACCP Guidelines

a steady state(50to60%inhibition)after4to7days. Such level of maximal inhibition was comparable to that achieved with ticlopidine,500mg/d,although the latter showed a slower onset of the antiplatelet effect than the clopidogrel.No appreciable differ-ences in the maximum inhibitory effects of50,75, and100mg of clopidogrel were noted in this study.230As would be expected from these pharma-cokinetic and pharmacodynamic features,a loading dose(eg,300mg)of clopidogrel results in a much more rapid onset of platelet inhibition than is achieved with the75-mg dose.236

Several recent studies have examined the ade-quacy of a300-mg loading dose of clopidogrel in patients scheduled for cardiac catheterization as potential candidates for PCI.237–240After loading with600mg of clopidogrel,the full antiplatelet effect of the drug was achieved after2h.237More-over,a loading dose of600mg resulted in higher plasma concentrations of the active metabolite,clo-pidogrel,and the carboxyl metabolite than did a loading dose of300mg.238ADP-induced platelet aggregation also was significantly lower in patients receiving600mg than in those receiving300 mg.238–240The incremental antiplatelet effect of 900mg over600mg of clopidogrel appears mar-ginal,238,239possibly because of limited drug ab-sorption.238

Clopidogrel treatment exhibited marked interindi-vidual variability in inhibiting platelet function in three different studies241–243of patients undergoing elective PCI and stenting.A variable proportion of these patients were considered to be clopidogrel nonresponders or to have clopidogrel resistance based on ADP-induced platelet aggregation.Three separate studies241,244,245suggested that concurrent treatment with lipophilic statins that are substrates of CYP3A4(eg,atorvastatin and simvastatin)may inter-fere with the inhibitory effects of clopidogrel on platelet function.In the study of Lau et al,244 atorvastatin,but not pravastatin,attenuated the an-tiplatelet effect of clopidogrel in a dose-dependent manner.Because many drugs are metabolized by CYP3A4,246it is likely that other drugs may modify the systemic bioavailability of the active metabolite of clopidogrel and affect its clinical efficacy.More-over,variable metabolic activity of CYP3A4may contribute to the interindividual variability in the platelet inhibitory effects of clopidogrel,as noted above.Although ex vivo measurements of ADP-in-duced platelet aggregation have suggested a pharma-cokinetic interaction between a CYP3A4-metabolized statin and clopidogrel,post hoc analyses of placebo-controlled studies247,248of clopidogrel,a single center cohort study,249and data from a large multinational registry250have failed to detect a statistically signifi-cant clinical interaction between the two.However, it should be emphasized that retrospective post hoc analyses have limitations that preclude definitive conclusions.Moreover,the lack of information on statin daily doses used in trials notably restricts our ability to assess the dose dependence of potential drug interactions.

As with aspirin,both the mechanism(s)and the clinical relevance of clopidogrel resistance or nonre-sponsiveness251remain to be established.Thus,no test of platelet function can currently be recom-mended to assess the effects of clopidogrel in the individual patient,as there is no uniformly estab-lished method for quantification of ex vivo platelet reactivity after clopidogrel treatment and to what extent platelet activity is inhibited by the drug.251 Thus,the active metabolite of clopidogrel has a pharmacodynamic pattern quite similar to that of aspirin in causing cumulative inhibition of platelet function on repeated daily administration of low doses.As in the case of aspirin,platelet function returns to normal7days after the last dose of clopidogrel.Both the cumulative nature of the in-hibitory effects and the slow rate of recovery of platelet function are consistent with the active moi-eties of aspirin(acetylsalicylic acid)and clopidogrel (active metabolite),causing a permanent defect in a platelet protein that cannot be repaired during the 24-h dosing interval and can only be replaced as a function of platelet turnover.This consideration also justifies the once-daily regimen of both drugs,de-spite their short half-life in the human circulation.It should be noted,however,that although aspirin currently is used at doses that represent a2.5-to 10-fold excess over the dose of30mg necessary and sufficient to fully inactivate platelet COX-1 activity on repeated daily dosing,25,62clopidogrel is used at doses causing only partial inactivation of P2Y12.Thus,the main determinants of the inter-individual variability in the antiplatelet effects of the two drugs are substantially different(Table6). Bleeding time measurements performed in the same multiple dose study230described above showed a comparable prolongation(by1.5-to2.0-fold over control)at50to100mg/d of clopidogrel or500mg/d ticlopidine.

Clopidogrel has undergone a quite unusual clinical development,with very limited phase II studies and a single,very large phase III trial(ie,CAPRIE,to test its efficacy and safety at75mg/d vs aspirin at325 mg/d).231CAPRIE is unique among the studies that have directly compared antiplatelet agents against aspirin in that it incorporated three groups of pa-tients,all of whom are recognized to be at an increased risk of recurrent ischemic events:those who have experienced a recent stroke or recent MI

https://www.360docs.net/doc/b711003600.html, CHEST/133/6/JUNE,2008SUPPLEMENT217S

(美国胸科医师学会)ACCP第三版指南精要之肺部小结节的评估

ACCP第三版指南精要之肺部小结节的评估 南京军区南京总医院呼吸内科姚艳雯宋勇 肺癌是目前致死率第一位的肿瘤[1]。究其原因,许多病人在诊断已发生转移是其中一重要因素。若能在早期即进行诊断,必定能降低肺癌的死亡率[2]。而肿瘤的最初起病可能就只表现为肺部的结节,临床医生或患者本身稍一疏忽,可能便会延迟肺癌的诊断。在临床中,我们不难发现,体检发现的肺部小结节并不少见,但这样的肺部结节是否是肺癌?如何对肺部结节进行评估?这是临床上的难点也是重点[2]。最新的第三版ACCP(美国胸科医师学会)指南,单独分出一部分来详述肺部结节的评估,由此为临床应用提供很大帮助[3]。 目前国内外公认的肺部结节的定义是:小的、成灶的、圆形、实质或混合性的影像学不透明影[4]。其中一部分为孤立性肺结节:单一的、边界清楚的、影像不透明的、直径小于或等于3cm、周围完全由含气肺组织所包绕的病变,没有肺不张、肺门增大或胸腔积液表现的肺部结节[5]。直径大于3cm的肺部局灶性改变被称为肺部团块,并且被认为是支气管源性的肿瘤可能大,因讨论的较多,这里略去不谈[6]。另外,因弥漫性的肺部结节或大于10个的肺部结节通常伴随着症状,并提示着肺外恶性疾病的转移或急性感染或炎症,因此也不再进一步讨论[3, 7]。此篇主要讨论的是无症状的肺部结节的评估。 ACCP指南所提出的肺部结节的评估全部是针对无法确定良恶性的结节[3]。因为直径小于等于8mm的结节恶性程度相对较低,因此将这部分结节从肺部结节中单独分类出来。另外就性质而言,实质性结节和亚实质性结节也分类开来,亚实质性结节进一步被分为纯磨玻璃样影或部分实质混合。在评估病人的肺部结节时,应将重点放在结节的大小、形态及恶性疾病的危险因素和是否合适进行后续治疗这些方面。 当胸部X平片或胸部CT提示可见性的不明性质的结节时,应首先回顾该病人的既往影像学资料[8]。如果影像学提示不明性质的实质性结节稳定2年以上,则不需做进一步的诊断检查。若该结节为胸部X平片所发现,则需要作进一步的胸部CT检查,以帮助明确结节性质[9]。 大于8mm的实质性结节 根据先前所述的肺部结节分类,对大于8mm的实质性结节,临床医生应对结节的检测前恶性概率进行评价,或者通过临床判断进行定性分析,或者通过经过认证的模型进行定量分析[3, 10]。当评估认为大于8mm的实质性结节的恶性概率低(5%-65%)时,可进行功能影像检查,推荐PET,来定义结节的性质。而对恶性概率较高(>65%)的大于8mm的实质性结节而言,可利用PET进行恶性结节的治疗前分期,但不能通过功能影像分析结节特征,此时需要临床医生对后续的不同诊断策略的获益和风险进行权衡,并且要注重患者的本身意愿[9]。 后续处理策略主要包括三个部分:CT扫描监测、非手术性的活检、外科诊断[3, 4, 11]。对于大于8mm 的实质性结节而言,CT扫描监测将应用于以下情况:1.临床恶性概率很低(<5%)时;2.临床恶性概率低(30%-40%以下),并且功能影像阴性(如,PET上代谢不高或动态增强CT上增强不超过15个hounsfield 单位)提示检测后恶性概率很低时;3.细针活检无法诊断并且PET上代谢不高时;4.向患者全面告知病情后患者自己选择该项非侵入性处理方案时。以上情况下,CT扫描监测应选用非增强的低剂量扫描,频率在3到6个月,9到12个月及18到24个月时[12]。当结节在监测扫描时提示具有明显的恶性增长时,除非特殊紧急,否则需要考虑行非手术性的活检和/或外科手术切除以帮助诊断[13, 14]。

美国最佳医生心胸外科CraigJ.Baker.

美国心胸外科最佳医生 Craig J. Baker医学博士 克雷格·贝克医学博士(Dr.Craig J. Baker,MD),现任洛杉矶县南加州大学医学中心(LA County USC Medical Center)心脏外科主任医师,并兼任南加州大学凯克医学院(USC, Keck School of Medicine)的临床外科教授。他于杜兰大学人文科学学院(Tulane University, College of Arts and Sciences)获得生物学学士学位,继而进入华盛顿乔治城大学医学院(Georgetown University, School of Medicine)获得医学博士学位,毕业后在南加州大学凯克医学院实习,完成了长达7年的医学训练,其中包括3年的心胸外科住院医师训练。此外,他还在在洛杉矶儿童医院(CHLA)完成了儿童心胸外科专科临床培训。 贝克教授擅长心胸外科手术、心血管疾病、胸外科手术,并专精于儿童和成人先天性心脏病(Pediatric And Adult Congenital cardiac Disease)、心脏移植(Cardiac Transplantation)、冠状动脉搭桥术,(Coronary Artery Bypass Grafting)、心脏瓣膜修复和更换(Valve Repair And Replacement),以及所有类型的复杂成人心脏手术(包括微创心脏瓣膜手术)等领域。

贝克教授是美国普通外科医学委员会(American Board of General Surgery)和美国胸外科医学委员会(American Board of Thoracic Surgery)双重认证的专科医师,亦是包括胸外科医师协会(Society of Thoracic Surgeons)在内的多个医学专业组织协会的资深研究员。他在成人和小儿心脏手术方面发表了大量的同行审阅性质医学论文和教科书章节,为许多权威医学专业期刊杂志和医学院所收录。 贝克教授目前执业于南加州大学医院(USC University Hospital)、洛杉矶儿童医院(Childrens Hospital Los Angeles)和南加州大学医疗保健咨询中心-2(USC Healthcare Consultation Center II)。临床治疗包括:先天性心脏病、心脏移植、冠状动脉旁路移植(冠脉搭桥手术)、心脏瓣膜修复和更换、微创瓣膜手术、复杂的成人心脏手术(所有类型)。 贝克教授于2012年获得美国医学界最高奖项之一的“卡斯尔康诺利最佳医师奖”(Castle Connolly's Top Doctors ),他在心脏外科手术领域拥有近20年临床经验,代表了当前世界级心脏外科手术的一流水平。

17-抗凝或抗血小板药物治疗患者接受区域麻醉与镇痛管理的专家共识(2017)

抗凝或抗血小板药物治疗患者接受区域麻醉与镇痛管理的专家共识 (2017) 王秀丽王庚(共同执笔人)冯泽国江伟李军张兰陈绍辉金善良袁红斌徐懋(共同执笔人)郭向阳(共同负责人) 止血机制正常的患者,区域麻醉导致封闭腔隙内(颅内、眼内或椎管内等)血肿和神经压迫损伤等严重并发症的概率很低。但对于使用抗凝、抗血小板等抗血栓药和其他导致止血异常的患者(创伤、大量失血、肝功能异常和DIC等),区域麻醉导致血肿的风险增加。一旦发生椎管内血肿或其它深部血肿,可能造成严重的不良后果,如截瘫、神经损伤、明显失血、气道梗阻等。 近年来,随着心脑血管疾病发病率的升高,服用抗血栓药物预防血栓的患者也日益增多。房颤、静脉血栓、机械瓣膜置换术后、冠脉支架置入术后等患者,通常会应用抗血栓药。此类患者接受区域麻醉时,其止血功能的异常增加了区域麻醉的风险。 对于围术期使用抗血栓药的患者,区域麻醉时机的选择很重要。麻醉科医师应该掌握常用抗血栓药的基本特点,选择合适的时机,将该类患者应用区域麻醉的风险降到最低。不同的患者,药物的代谢亦不同,应注意凝血功能的检查。熟悉抗血栓药物的药理特点并结合凝血功能检查,有助于麻醉科医师做出更加合理的选择。 本专家共识参考国内外相关指南,结合心脑血管及深静脉血栓用药和区域麻醉的应用,为抗凝或抗血小板药物治疗患者的区域阻滞管理提供意见,规范围术期的相应管理,供麻醉科医师和相关医务人员参考。 一、常用抗血栓药的基本药理 临床常用抗血栓药可分为抗凝血酶药、抗血小板药及纤维蛋白溶解药等。中草药和抗抑郁药也有改变凝血功能的作用。各类药物的基本药理作用简述如下。 (一)抗凝血酶药 1.间接凝血酶抑制剂

美国胸科医师协会第九版抗栓治疗及血栓预防指南静脉血栓栓塞性疾病最新进展

美国胸科医师协会第九版抗栓治疗及血栓预防指南静脉血栓栓塞性疾病最新进展 周玉杰, 杨士伟(首都医科大学附属北京安贞医院 心内科12病房,北京 100029) 通讯作者:周玉杰 E-mail:jackydang@https://www.360docs.net/doc/b711003600.html, 静脉血栓栓塞性疾病(venous?thromboembolism ,VTE )包括深静脉血栓(deep?venous?thrombosis ,DVT )和肺栓塞(pulmonary?embolism ,PE )。近日,美国胸科医师协会(American?College?of?Chest?Physicians ,ACCP )在Chest 杂志上公布了第9版《抗栓治疗及血栓预防指南》(ACCP-9)[1-4]。此版指南在第8版基础上,结合最新循证医学证据,对抗栓治疗进行了全面细致的推荐。本文将结合ACCP-9对VTE 的最新进展进行阐述。1 病因与诱因 Virchow 于1856年提出了导致静脉血栓形成的三因素假说:静脉血流淤滞、血管损伤和高凝状态。经过大量试验证实,该假说目前已得到了公认。许多参与上述过程的临床情况均可诱发静脉血栓形成,主要包括: (1)肢体制动:各种疾病导致的长时间卧床或肢体制动,如急性心肌梗死、主要脏器功能衰竭、脑卒中、大型手术后等; (2)静脉受压:如股动脉或静脉穿刺术后压迫止血导致下肢静脉回流受阻,妊娠、腹腔巨大肿瘤或大量腹水导致盆腔静脉或下腔静脉回流受阻等;(3)静脉曲张:主要是指深静脉曲张,浅静脉曲张一般不会导致血栓形成; (4)静脉损伤:如外伤或手术累及静脉、静脉穿刺、静脉炎等; (5)高凝状态:各种遗传性或获得性血栓形成倾向,如抗凝因子缺乏(抗凝血酶Ⅲ、C 蛋白或S 蛋白最常见)、弥漫性血管内凝血(disseminated? intravascular?coagulation ,DIC )、抗磷脂综合征、肾病综合征、恶性肿瘤、大量呕吐或腹泻导致的脱水、药物因素(长期口服避孕药、激素或抗肿瘤药物)等; (6)其他:如高龄、吸烟、肥胖等。2 病理与病理生理 以上因素往往不是孤立的,而是多种因素同时存在,相互影响,有时很难明确始动因素,但静脉损伤通常是导致血栓形成的关键环节。内膜损伤以后迅速激活内源性和(或)外源性凝血途径,在损伤部位形成血栓(thrombosis )。 血栓的起始部(即头部)主要由血小板及少量纤维蛋白构成,称为白血栓(pale?thrombus );血栓头部形成以后,下游血流缓慢和出现涡流,此时除血小板聚集以外,还有大量红细胞填充在纤维蛋白形成的网状结构内,称为混合血栓(mixed?thrombus );当混合血栓阻塞管腔以后,远端血流完全停止,血液凝固,此时血栓主要由纤维蛋白和红细胞构成,称为红血栓(red?thrombus )。白血栓与血管壁结合紧密,不易脱落;而红血栓与血管壁黏附力差,容易脱落,形成栓塞(embolism )。血液中存在凝血系统与抗凝血系统,凝血系统激活的同时抗凝血系统也被激活。因此在早期,血栓形成与溶解是一个动态过程,这一阶段血栓也最容易脱落而导致栓塞。如果促凝因素持续存在或血栓溶解不完全,未脱落的血栓可发生机化,血栓被肉芽组织取代。较大的血栓约2周可以完全机化,此时,血栓与血管壁紧密结合不易脱落,栓塞危险显著降

美国儿科学会母乳喂养和母乳使用的政策陈述

美国儿科学会关于母乳喂养和母乳使用的政策陈述 (武汉市疾病预防控制中心) 摘要:母乳喂养和人奶是婴儿喂养和营养供给的标准规范。因为母乳喂养在神经系统发育及其他的健康方面具有短期和长期的优势,所以婴儿营养应该是一项公共卫生的问题,而不是单纯的喂养方式的选择。 美国儿科学会重申:推荐6个月的纯母乳喂养,6个月添加辅食后继续母乳喂养,并可根据母婴双方的需要喂养至一年或一年以上。 母乳喂养在医学上的禁忌症很少。婴儿的生长发育应参考世界卫生组织(the World Health Organization,WHO)生长发育标准曲线,避免对婴儿低体重或发育迟缓的误诊。 医疗单位需根据美国儿科学认可的WHO/UNICEF“母乳喂养成功实现十步骤”来鼓励和支持初始的和持续的母乳喂养。美国卫生总署(US Surgeon General)的“行动呼吁”计划、美国疾病预防与控制中心(CDC)和美国医疗机构评鉴联合会(The Joint Commission)所支持的国家战略中,也涉及到在美国医院和社区中推广母乳喂养方面的行动。 儿科医生作为母乳喂养的倡导者在其执业过程和社区活动中对母乳喂养起到了重 要作用,所以儿科医生应知道非母乳喂养的健康风险,母乳喂养在社会经济上的益处,以及实现和支持母乳喂养所需要的技能。“职场母乳喂养”详细讲述了职场妈妈怎样维持哺乳喂养,以及支持母乳喂养对雇主们的好处。 关键词:母乳喂养;辅食添加;婴儿营养;哺乳;母乳;护理 缩略词:AAP——美国儿科学会 AHRQ——美国医疗保健研究与质量局 CDC——疾病预防控制中心 CI——可信区间 CMV——巨细胞病毒 DHA——二十二碳六烯酸 NEC——坏死性小肠结肠炎 OR——比值比

金贝口服液有望挽救肺纤维化患者

金贝口服液有望挽救肺纤维化患者 2019年12月18日济南日报记者刘文忠 发病率近年来不断上升的特发性肺纤维化(业内英文简称IPF),诊断后的平均生存期只有2.8年,不可逆+高死亡率,让它成了医学界公认的“不是癌症的癌症”。 服用进口西药,价格昂贵,毒副作用多,患者认可度不高;肺移植手术又面临供体、高额费用等问题。 中医药能否为治疗这种罕见病提供新的解决方案?传承精华,守正创新,来自济南的名老中医和制药企业宏济堂制药集团,正在加快技术攻关,组织有益尝试。 病因不明的“肺炎” “医生,我最近总是干咳,稍微活动一下,就感觉要窒息一样,喘不过气来。你给我看看,这是得了啥病了?”在某医院呼吸科,一位老年患者向医生诉说着自己的症状。 经过进一步观察、询问,这位患者不仅有干咳、呼吸困难、呼吸急促等外部表征,而且经常感到疲倦乏力,食欲不振,体重下降;手指、足趾末端还出现增生、肥厚现象,呈杵状膨大。 “经验不足的医生,很可能把它当成普通肺炎来治疗。因为从影像学检查或组织病理看,这些症状都是普通间质性肺炎的表现。”宏济堂中医院外聘专家陶凯教授说,“事实上,这是一种好发于中老年人的慢性、进行性、纤维化性间质性肺疾病,医学上叫做特发性肺纤维化(IPF)。” 国际上至今尚未查明IPF病因,其发病机制亦未完全阐明。医学界普遍认为,足够证据表明IPF与免疫炎症损伤有关,吸入物刺激与特定的遗传因素会增加IPF的患病风险,发病率男性高于女性,发病年龄通常在50-70岁之间。绝大多数IPF患者早期临床表现不典型,漏诊和延误诊断现象普遍,也易被误诊为慢阻肺、哮喘和充血性心力衰竭或其他肺部疾病。 2000年,美国胸科学会、欧洲呼吸学会、美国胸科医师协会首次确认IPF为一种独立的临床疾病。去年5月国家卫健委等5部门联合制定的《第一批罕见病

美国意思协会-CKD指南

Screening,Monitoring,and Treatment of Stage 1to 3Chronic Kidney Disease:A Clinical Practice Guideline From the American College of Physicians Amir Qaseem,MD,PhD,MHA;Robert H.Hopkins Jr.,MD;Donna E.Sweet,MD;Melissa Starkey,PhD;and Paul Shekelle,MD,PhD,for the Clinical Guidelines Committee of the American College of Physicians* Description:The American College of Physicians (ACP)developed this guideline to present the evidence and provide clinical recom-mendations on the screening,monitoring,and treatment of adults with stage 1to 3chronic kidney disease. Methods:This guideline is based on a systematic evidence review evaluating the published literature on this topic from 1985through November 2011that was identified by using MEDLINE and the Cochrane Database of Systematic Reviews.Searches were limited to English-language publications.The clinical outcomes evaluated for this guideline included all-cause mortality,cardiovascular mor-tality,myocardial infarction,stroke,chronic heart failure,composite vascular outcomes,composite renal outcomes,end-stage renal dis-ease,quality of life,physical function,and activities of daily living.This guideline grades the evidence and recommendations by using ACP’s clinical practice guidelines grading system. Recommendation 1:ACP recommends against screening for chronic kidney disease in asymptomatic adults without risk factors for chronic kidney disease.(Grade:weak recommendation,low-quality evidence) Recommendation 2:ACP recommends against testing for protein-uria in adults with or without diabetes who are currently taking an angiotensin-converting enzyme inhibitor or an angiotensin II–receptor blocker.(Grade:weak recommendation,low-quality evidence) Recommendation 3:ACP recommends that clinicians select phar-macologic therapy that includes either an angiotensin-converting enzyme inhibitor (moderate-quality evidence)or an angiotensin II–receptor blocker (high-quality evidence)in patients with hyperten-sion and stage 1to 3chronic kidney disease.(Grade:strong recommendation) Recommendation 4:ACP recommends that clinicians choose statin therapy to manage elevated low-density lipoprotein in patients with stage 1to 3chronic kidney disease.(Grade:strong recommenda-tion,moderate-quality evidence) Ann Intern Med.2013;159:https://www.360docs.net/doc/b711003600.html, For author affiliations,see end of text. This article was published online first at https://www.360docs.net/doc/b711003600.html, on 22October 2013. C hronic kidney disease (CKD)is nearly always asymp-tomatic in its early stages (1).The most commonly accepted de?nition of CKD was developed by Kidney Dis-ease:Improving Global Outcomes (KDIGO)(2)and the Kidney Disease Outcomes Quality Initiative (KDOQI)(3)as abnormalities of kidney structure or function,present for more than 3months,with implications for health.Cri-teria for CKD include markers of kidney damage (albu-minuria,as indicated by an albumin excretion rate of 30mg/24h or greater and an albumin–creatinine ratio of 3mg/mmol or greater [?30mg/g]);urine sediment abnor-malities;electrolyte and other abnormalities due to tubular disorders;abnormalities detected by histologic examina-tion;structural abnormalities detected by imaging;history of kidney transplantation or presence of kidney damage;or kidney dysfunction that persists for 3or more months,as shown by structural and functional abnormalities (most often based on increased albuminuria,as indicated by a urinary albumin–creatinine ratio of 3mg/mmol or greater [?30mg/g])or a glomerular ?ltration rate (GFR)less than 60mL/min/1.73m 2for 3or more months. Traditionally,CKD is categorized into 5stages that are based on disease severity de?ned by GFR (3)(Table 1);stages 1to 3are considered to be early-stage CKD.People with early stages of the disease are typically asymptomatic,and the diagnosis is made by using laboratory tests or im-aging.In 2013,KDIGO revised CKD staging to consider both 5stages of GFR as well as 3categories of albuminuria to de?ne CKD severity (2). *This paper,written by Amir Qaseem,MD,PhD,MHA;Robert H.Hopkins Jr.,MD;Donna E.Sweet,MD;Melissa Starkey,PhD;and Paul Shekelle,MD,PhD,was developed for the Clinical Guidelines Committee of the American College of Physicians.Individuals who served on the Clinical Guidelines Committee from initiation of the project until its approval were Paul Shekelle,MD,PhD (Chair );Roger Chou,MD;Molly Cooke,MD;Paul Dallas,MD;Thomas D.Denberg,MD,PhD;Nick Fitterman,MD;Mary Ann Forciea,MD;Robert H.Hopkins Jr.,MD;Linda L.Humphrey,MD,MPH;Tanveer P.Mir,MD;Douglas K.Owens,MD,MS;Holger J.Schu ¨nemann,MD,PhD;Donna E.Sweet,MD;and Timothy Wilt,MD,MPH.Approved by the ACP Board of Regents on 17November 2012. See also: Print Summary for Patients.......................I-28Web-Only CME quiz

美国外科住院医师培养简介

?继续教育?美国外科住院医师培养简介 顾晋 刘玉村 由美国中华医学基金会(China medical board,C M B)资助的北京医科大学医学教育考察团于1999年10月对美国多所大学医学院进行了为期3个月的考察,对较有代表性的哥伦比亚大学内外科学院(C olumbia University C ollege of Physicians&Surgeons)有了较为深刻的了解。作为外科医师,仅对外科住院医师的培养及外科各专业设置情况作一介绍。 美国医学教育制度及医学生 毕业后的职业选择 在美国,几乎所有正规医学院的报考者必须己经完成普通高等教育的本科4年学习。经过严格考试被录取的考生需再花4年时间完成医学院的教育。第1,2年学习医学基础理论;第3年开始进入临床阶段学习;到第4年的医学生主要进行各科临床实习。学生要根据自己的特点、能力、兴趣选择专科实习,此时的医学生就开始考虑毕业后的工作选择并寻找毕业后作住院医师的用人单位。由于每年各医院及大学所能提供外科住院医师的位置相当有限,特别是有名的医学中心。因此,立志成为外科医师的学生将面临激烈的竞争。每年10月,美国国家住院医师选拔计划(national resi2 dent match program,NRMP)要公布全美各地各大医院及医学中心该年度所接受住院医的科室及名额。学生应向NRMP提出申请,然后再经过用人部门的严格考试和面试后确定下一年度准备接受的新住院医师的名单。 外科住院医师的培训 一、住院医师的轮转计划 作者单位:100036北京医科大学临床肿瘤学院外科(顾晋);北京医科大学第一临床学院外科(刘玉村) 医学院毕业生已经取得外科住院医 师资格后,开始他们为期五年的培训计 划。医学院毕业后的第1年住院医师称 为Intern,第2年的住院医师称为Junior resident,第3、4年的住院医师称为Senior resident,到第5年则为住院总医师(Chief resident)。这个阶段是住院医师的最后1 年的训练。 二、各年住院医师的职责 美国外科职业手册(the surgical ca2 reer handbook)中对外科医师的职责做了 较详细的规定:“当你作为一个外科医 师,就意味着你是整个医疗过程的主体, 意味着你必须提供给患者所需要的一 切:术前诊断、术中操作、术后随访及治 疗”。 1.住院医师的监督制度:正在接受 住院医师培训的医师,必须接受上级医 师的监督。尽管高年住院医师已经具备 了一定的临床经验,也必须接受主治医 师(attending surgeon)的监督,强调逐级监 督的责任制度。对患者的管理最主要的 是主治医师负责。主治医师根据每个住 院医师的能力、知识水平与技术熟练程 度安排适合的临床工作。 2.普通外科的训练(第1、2年):普 通外科的训练是外科住院医师培训的基 础。头2年训练主要是外科基本技能, 直接接受主治医师的指导。第1年的住 院医师必须经过几个月在普外科轮转, 规定的轮转科室应包括:消化内窥镜室、 烧伤科、普外门诊和器官移植科。第1、 2年的住院医师比较重点强调外科患者 的病史采集、体格检查、基本手术的手术 前术后处理、基本手术操作及外科适应 证的选择。在急诊外科轮转应学习休克 的治疗、常见骨折的处理、伤口缝合、外 科抗生素的使用、输血的适应证等。在 第1、2年的住院医师培训阶段,在有经 验的主治医师的指导下完成较简单的手 术。住院医师应每天参加查房,并学习 如何正确地与患者接触;在各级查房中, 汇报病史、讲述并分析X线片及CT检 查以及化验结果、提出可能的诊断和治 疗意见。 3.高年住院医师培训:第3年住院 医师重点要进行外科如胸科、骨科、泌尿 科等各专业的训练,他们在选择的专业 领域里进行专科技术培养。第3年住院 医师的责任比普通外科住院医师范围有 所扩大,要负责住院患者,门诊患者和急 诊患者的管理,并且负责院内会诊。在 有经验的主治医师的指导下完成手术。 同时也协助指导低年住院医师,并担任 医学院3年级的课程教学工作。 4.第4年住院医师:参加更多的手 术,指导初年住院医师和临床教学工作。 可以相对独立地管理患者,特别是住院 患者和重症监护患者,有些可以有机会 进行专业化的训练,包括激光治疗、腹腔 镜和纤维内窥镜的操作。 5.科学研究:所有的医师培训中心 均有实验研究的内容。根据计划,住院 医师在挑选培养计划时就已经确定了他 的科研题目,所选的研究课题可以是临 床的应用,也可以是基础理论的研究。 住院医师可以参加整个课题组的研究, 或单独完成独立的研究课题。 6.住院总医师(第5年):住院总医 师是住院医师培训的最后1年,他可以 在这1年里独立地完成主要的外科手 术,并对患者术前及术后观察和处理负 责。组织外科查房和各科间的联合查 房。此外,还要担任医学生的教学工作 并指导低年住院医师工作。 外科各专业亚科的设置 在经过5年的外科基本训练的住院 医师在第6年开始面临着又一个选择, 即外科专业的选择。5年的外科基本训 练使他们具备了外科专业的基础知识和 基本技能,可以进一步接受专科训练。

第06章 美国内科医师协会:成人急性呼吸道感染抗菌药物指导

美国内科医师协会:成人急性呼吸道感染抗菌药物指南 急性呼吸道感染(ARTI)是呼吸科门急诊就诊的最常见病因之一,对于成人患者常会开具抗菌药物。ARTI 包括急性单纯性支气管炎、咽炎、鼻窦炎和普通感冒。对ARTI 患者不适当的使用抗菌药物不仅会导致耐药,还会给公共卫生带来严重威胁。 为控制ARTI 抗菌药物的滥用情况,合理控制由此导致的耐药菌株感染发病率增加和药物使用支出,近日美国内科医师协会(ACP)和美国疾病预防控制中心联合发布了成人急性呼吸道感染抗菌药物使用指南,旨在提供高质量的诊疗建议。 急性单纯性支气管炎 急性单纯性支气管炎是一种大气道(支气管)自限性炎症,伴有咳嗽,可持续6 周。咳嗽(可出现或不出现)同时伴有轻微的全身症状。急性支气管炎是最常见的门诊成人患者诊断,在美国有70% 的患者会使用抗菌药物。急性支气管炎导致的不合理抗菌药物使用在成人中明显多于其他ARTI 综合征。 1. 判断细菌感染的可能性 90% 表现为咳嗽的门诊患者是由于病毒感染所致,其他非病毒病原体如肺炎支原体、肺炎衣原体可偶尔导致急性支气管炎。百日咳博德特氏菌如果存在社区流行也应该加以考虑。然而判断究竟是否为病毒感染是非常困难的。 出现脓痰或者痰的颜色改变(如黄色、绿色)并不意味着细菌感染。脓痰的出现是由于炎症细胞或者脱落的粘膜上皮细胞。 对于70 岁以下免疫功能正常的健康成人,符合下列标准时才需要考虑肺炎:心动过速(> 100 次/ 分),呼吸加快(>24 次/ 分)、发热(口温>38℃)及胸部检查的异常(啰音、羊鸣音、触觉语颤)。 2. 管理策略 大部分近期指南对于急性单纯性支气管炎都不建议常规使用抗菌药物,除非是出现了肺炎。 使用对症处理的药物可能会使患者受益,这些药物包括镇咳药(右美沙芬、可待因)、祛痰药(愈创甘油醚)、第一代抗组胺药(苯海拉明)、减充血药(去氧肾上腺素)、β 激动剂(沙丁胺醇)等,但是这些治疗手段的证据资料有限。 3. 高水平建议(指证据等级) 除非怀疑存在肺炎,否则不应经验性使用抗菌药物。 咽炎

抗凝及抗血小板药物应用及护理

抗凝及抗血小板药物应用及护理 抗凝药是指能降低机体的凝血功能,防止血栓形成或对已形成血栓的可防止其进一步发展的药物 抗凝药物的基本分类 1、阻止纤维蛋白形成的药物:肝素、低分子肝素、华法林、蚓激酶等 2、促进纤维蛋白溶解的药物:尿激酶、链激酶、降纤酶、阿替普酶等 3、抗血小板药物:阿司匹林、前列环素、双嘧达莫、氯吡格雷等 一、阻止纤维蛋白形成的药物 1、肝素类 成份及种类:肝素钠/钙,普通/低分子 机制: 在体内外都有抗凝血作用 与凝血酶原-III结合,对活化的Ⅱ、Ⅸ、Ⅹ、Ⅺ和Ⅻ凝血因子的抑制作用。 2香豆素类 华法令:为香豆素类口服抗凝剂,结构与VitK类似 机制:

竞争性拮抗VitK的作用,使维生素依赖的凝血因子作用产生障碍,致II、VII、IX、X失活,使凝血酶原时间延长。 对已经合成的凝血因子因子并无直接对抗作用。 华法林没有溶栓(化栓)的作用 药动学论 1、口服吸收迅速,生物利用度100%作用发挥慢 2、半衰期36-42小时 3、几乎完全通过肝脏代谢清除,主要通过肾脏排泄 4、剂量反应关系变异很大,因此需要严密监测INR 5、受遗传因素及环境因素的影响,小剂量华法林引起较高的出血并发症的发生 6、药为酸性,遇碱性药物则失去抗凝性能 小结—不宜合用的药物 1、解热镇痛药 2、广谱抗生素 3、镇静剂 4、同化激素、甲状腺素、肾上腺皮质激素 5、口服降糖药 6、抗结核药 7、制酸药、轻泻药 监测 1、用药的第一周每3天监测1次INR。

2、INR达到治疗目标并维持至少两天,1周监测1次 3、当INR稳定后对于门诊患者,可以每4周监测1次 INR目标值 多数75岁以下的房颤患者2.5(2.0-3.0),75岁以上出血高危患者2.0(1.6-2.5) 不良反应 1、常见的不良发应是出血,最常见是鼻出血,此外有齿龈,胃肠道、泌尿生殖系统等 2、对于出血,一般采用维生素K1对抗治疗 3、输入新鲜的血液。 二、促进纤维蛋白溶解的药物 尿激酶 1、来源:健康人尿液中提取的蛋白水解酶,也可由 2、机制:直接作用于血块表面的纤溶酶原,产生纤溶酶,使纤维蛋白溶解 注意事项 1、不可肌注给药。否则可发生红肿,静注后穿刺部位要加压。 2、现用现配。溶解后存放会失去活性,使药效降低。 3、本药稀释宜用近中性液体,避免酸性液体,易使之降解。 三、抗血小板药物

静脉血栓栓塞性疾病的抗血栓治疗—解读美国胸科医师学会循证医学临床实践指南(+第9+版)

·综述与讲座· 静脉血栓栓塞性疾病的抗血栓治疗———解读美国胸科医师学会循证医学临床实践指南(第9版) 李圣青(解放军第四军医大学西京医院呼吸与危重症医学科,陕西西安710032) 摘要:美国胸科医师学会(ACCP)抗栓治疗和血栓预防临床实践指南第9版的抗血栓治疗篇重点讲述了静脉血栓栓塞性疾病(VTE)的治疗。与第8版指南相比,第9版指南有很大程度的修改并增添了部分新的内容。除了抗血栓药物、使用装置或外科手术技术在深静脉血栓形成(DVT)和肺栓塞(PE)(统称为VTE)的使用建议外,还提供了关于血栓后综合征(PTS),慢性血栓栓塞性肺动脉高压(CTPH),偶然诊断的(无症状)DVT或PE,急性上肢深静脉血栓形成(UEDVT),浅静脉血栓形成(SVT),内脏静脉血栓形成和肝静脉血栓形成的治疗建议。本文对相关部分内容进行了解读。 关键词:静脉血栓栓塞症;急性下肢深静脉血栓;急性肺血栓栓塞;抗凝;溶栓 中图分类号:R543.6文献标志码:A doi:10.3969/j.issn.1671-3826.2013.06.41文章编号:1671-3826(2013)06-0647-04 美国胸科医师学会(ACCP)于2012年发布了第9版抗栓治疗和血栓预防临床实践指南[1],与第8版指南相比有较大程度的修改并增添了部分新的内容,且更为简洁明了。对待不同的治疗既表现出谨慎而稳妥,又表现出积极和开放。指南规定高质量的临床证据为A级,中等质量的临床证据为B级,低质量的临床证据为C级。根据临床证据等级的不同,将推荐等级分为强烈推荐(1级)和建议(2级)。强烈推荐适用于大多数病人,建议则根据患者的具体情况而定,包括患者的个人选择。绝大多数推荐和建议都是基于静脉血栓栓塞性疾病(VTE)的复发风险与抗凝或溶栓出血风险二者之间的权衡而得出的结论。 1急性深静脉血栓形成(DVT)的治疗 1.1急性DVT抗凝治疗的时机对于高度怀疑VTE患者抗凝时机的把握,第9版指南给出了更加积极的抗凝建议。对于临床高度怀疑急性VTE的患者,建议胃肠外抗凝治疗而不是等相关检查的结果回报后治疗(2C级)。对于临床中度怀疑急性VTE的患者,如果诊断检查的结果回报预计将超过4h,建议肠外抗凝剂治疗(2C级)。对于临床低度怀疑急性VTE的患者,如果诊断检查的结果预计在24h内回报,建议不使用肠外抗凝剂治疗(2C级)。 1.2急性孤立性下肢远端DVT的处理对于急性孤立性下肢远端DVT患者,第9版指南倾向于保守性的超声观察,而不是积极的抗凝治疗。指南建议如无严重症状或进展危险因素的患者,建议连续观察深静脉影像学变化2周而不进行初始抗凝治疗(2C级)。对于急性孤立性下肢远端DVT患者,伴有严重症状或具有进展危险因素的患者,建议初始抗凝治疗而不是连续观察深静脉影像学变化(2C 级)。对于连续观察影像学变化的急性孤立性下肢远端DVT 作者简介:李圣青(1970-),女,安徽芜湖人,副教授,副主任医师患者,如果血栓没有扩大,不推荐抗凝治疗(1B级);如果血栓扩大,但仍局限于远端静脉,推荐抗凝治疗(1C级);如果血栓延伸到近端静脉,推荐抗凝治疗(1B级)。 1.3急性DVT的抗凝治疗第9版指南中普通肝素(UFH)在急性DVT抗凝治疗中的地位显著下降。口服维生素K拮抗剂(VKA)的时机提升至与肠外抗凝药物同步使用,并积极鼓励家庭初始抗凝治疗。指南建议口服VKA 治疗下肢急性DVT时,推荐初始胃肠外抗凝治疗[低分子量肝素(LMWH)、磺达肝素、静脉注射UFH或皮下注射UFH],优于无初始治疗(1B级)[2]。对于急性下肢DVT患者,推荐早期使用VKA(如在肠外治疗的当天开始使用),优于延迟使用VKA。推荐连续肠外抗凝治疗至少5d,直到国际标准化比值(INR)达到2.0以上至少24h(1B级)。对于急性下肢DVT患者,建议LMWH或磺达肝素治疗,优于静脉(2C级)或皮下注射UFH治疗(LMWH2B级,磺达肝素2C级)。急性下肢DVT患者,如其家庭条件允许,推荐家庭治疗,优于在医院接受治疗(1B级)。 1.4急性DVT的滤器植入第9版指南对滤器植入的适应证要求更加严格。指南建议对于急性下肢DVT患者,应使用抗凝治疗,不推荐植入下腔静脉滤器(IVCF)(1B 级)[3]。对于急性下肢近端DVT,并有抗凝治疗禁忌证的患者,推荐植入IVCF(1B级)。对于急性下肢近端DVT和IVCF植入来替代抗凝治疗的患者,如其出血风险去除,建议常规抗凝(2B级)。 1.5急性DVT的其他治疗建议第9版指南对急性DVT 的导管溶栓和手术治疗的建议很保守,通常在抗凝效果不佳的情况下才考虑,并要求有丰富经验的专业团队才能进行上述操作。指南建议对于急性下肢近端DVT患者,单纯抗凝治疗,优于导管溶栓(CDT)治疗(2C级)。对于急性下肢近端DVT患者,建议单纯抗凝治疗,优于全身溶栓治疗

麻醉习题集美国麻醉医师协会(ASA)分级

麻醉习题集 1.有关美国麻醉医师协会(ASA)分级下列哪项正确 A、I 级病人体格健康,发育营养良好,重要器官功能正常 B、Ⅱ级病人重要器官轻度病变,但功能代偿健全 C、Ⅲ级病人重要器官功能受损,已丧失工作能力 D、Ⅳ级病人重要器官功能已失代偿,麻醉手术风险很大 E、Ⅴ级病人病情危重、濒临死亡,麻醉和手术异常危险 2.肥胖对生理功能的影响,下列哪项不对 A、使肺-胸顺应性降低 B、使肺活量、深吸气量减少 C、使功能余气量增加 D、使通气/血流比值失调 E、使肺泡通气量降低 3.下列哪项不能提示病人呼吸功能不全 A、肺活量低于预计值的60% B、通气储量百分比<70% C、第一秒用力呼气量与用力肺活量的百分比(FEV1/FVC%)<60%或50% D、呼气时间短于3 秒 E、屏气时间短于20 秒 4.有关麻醉前禁食禁饮哪项不正确 A、在应激情况下胃排空加速 B、成人择期手术前禁食12小时,禁饮4小时 C、小儿禁食4~8 小时,禁水2~3时 D、急症病人病情紧急时,不能等待禁食,应立即手术 E、饱胃病人必需立即手术时,应采取避免呕吐误吸的措施 5.有关高血压病人术前评估和准备哪项正确 A、术前宜使用中枢性降压药控制血压 B、术前一天应停用所有抗高血压药 C、术前血压应降至正常才能手术 D、收缩压升高比舒张压升高危害小 E、麻醉危险性主要决定于重要器官是否受累以及其受累的严重程度 6.麻醉前用药的目的不包括 A、清除病人紧张、焦虑及恐惧心情 B、预防术后感染 C、提高病人痛阈,缓解术前疼痛 D、抑制呼吸道腺体分泌功能 E、减轻或消除手术或麻醉引起的不良反射 7.麻醉前用药的基本原则下列叙述哪项正确

吲哚布芬(美国胸科协会抗血小板药物指南)

DOI 10.1378/chest.08-0672 2008;133;199S-233S Chest Carlo Patrono, Colin Baigent, Jack Hirsh and Gerald Roth * Antiplatelet Drugs ull.html https://www.360docs.net/doc/b711003600.html,/content/133/6_suppl/199S.f and services can be found online on the World Wide Web at: The online version of this article, along with updated information ISSN:0012-3692 )https://www.360docs.net/doc/b711003600.html,/site/misc/reprints.xhtml (of the copyright holder. may be reproduced or distributed without the prior written permission Northbrook, IL 60062. All rights reserved. No part of this article or PDF by the American College of Chest Physicians, 3300 Dundee Road, 2008Physicians. It has been published monthly since 1935. Copyright CHEST is the official journal of the American College of Chest

相关文档
最新文档