Inflammation and Immunity as Targets for Drug Therapy in Acute Coronary Syndrome




Inflammation has been associated with myocardial infarction, even before the recognition that acute coronary thrombi cause coronary occlusion and a time-dependent loss of myocardial muscle. In response to injury, the immune system plays a crucial role in clearing the necrotic cells and the matrix debris, which initiates the process of scar formation. Both the immune and the inflammatory systems act in concert to coordinate the migration of inflammatory cells into the injured myocardium. Inflammation is essential to heal the infarction. It has been hypothesized that expansion of the infarction is a common maladaptive process, as is the case of reperfusion injury. Preclinical and clinical research on reperfusion injury has been a major focus over the past two decades. Controversy exists, however, as to whether and when reperfusion injury plays an important role in clinical myocardial infarction, and the modulation of the inflammatory response in the hours following an acute coronary syndromes has not resulted in demonstrable clinical benefits. Animal models of reperfusion injury show that the spontaneous reperfusion of recently infarcted myocardium triggers several intense inflammatory signals that culminate in excessive cell death and adverse myocardial remodeling. Inflammation is a major component of reperfusion injury and several adjunctive anti-inflammatory strategies have been tested in clinical trials. This chapter will focus on clinical investigation targeting the inflammatory and the immune system in patients with recent myocardial infarction.


The Adverse Contribution of Inflammation during the Reperfusion Injury


Although aggressive revascularization and optimal medical therapy have resulted in improved outcome, mortality and morbidity remain high following acute myocardial infarction (AMI). In a contemporary cohort of patients with AMI, the in-hospital mortality ranged from 7% in the contemporary international Global Registry of Acute Coronary Events (GRACE) up to 28% in the MONICA Project. To date, the only intervention proven to reduce infarct size is early revascularization, either through fibrinolytic therapy or primary percutaneous coronary intervention (PCI). Although restoration of blood flow to the infarcting myocardium is essential to prevent irreversible injury, reperfusion itself may worsen tissue injury in excess of what would be produced by ischemia alone. The observation in randomized clinical trials of fibrinolytic therapy versus control of excess mortality the first day following fibrinolytic therapy may reflect this phenomenon. This second wave of damages, called the reperfusion injury, has been thought to paradoxically aggravate the initial damages caused by ischemia.


If reperfusion injury is occurring in human myocardial infarction, the total benefit of a reperfusion (the myocardial salvage) can be expressed as the diminution of the necrotic zone due to timely reperfusion minus the paradoxical damage caused by reestablishment of flow to the injured myocardium (the reperfusion injury). The reperfusion injury, in return, can be broken down into two major components: vascular reperfusion injury and cellular reperfusion injury. Vascular reperfusion injury results in the “no reflow” seen when the infarct-related epicardial artery is reopened but coronary flow to the downstream myocardium is not re-established. The vascular injury is assumed to be caused by capillary plugs made of polymorphonuclear leukocytes (PMNs) and inflammatory microthrombi with distal embolization. Cellular reperfusion injury is a complex phenomenon that involves several defective pathways, among which the acute inflammatory response plays an important role. This cell injury results from the release of reactive oxygen radicals, intracellular calcium overload, the opening of the mitochondrial permeability transition pores, and apoptosis. The acute inflammatory response has also been divided into distinct components: the activation of the complement cascade, the intramyocardial infiltration by PMN leukocytes, and the toll-like receptor mediated pathways. The principal components of the reperfusion injury are summarized in Table 25-1 .



TABLE 25–1

The Components of the Reperfusion Injury


















Total myocardial salvage = [Diminution of necrotic core] − [reperfusion injury]
Reperfusion injury = [Vascular reperfusion injury] + [cellular reperfusion injury] + [apoptosis]
Vascular reperfusion injury = [Inflammatory cells capillary plugging] + [inflammatory microthrombi] + [distal embolization]
Cellular reperfusion injury = [Reactive oxygen radicals] + [intracellular calcium overload] + [opening of the mitochondrial permeability transition pores] + [acute inflammatory response]
Acute inflammatory response = [Complement system activation] + [toll-like receptor-mediated pathways] [polymorphonuclear leukocyte infiltration]


There are several comprehensive reviews on the mechanisms of reperfusion injury and on interventions targeting the potential mediators of cellular or “lethal” reperfusion injury. , Likewise, strategies used to reduce the inflammation in the coronary vasculature can be reviewed in the chapter on plaque passivation (see Chapter 26 ). This chapter will focus on drugs and interventions that specifically target the inflammation and immunity processes occurring in the myocardium after an acute coronary syndrome. A detailed understanding of the role played by the inflammatory and immune systems is required before exploring the overall disappointing clinical experience seen with many cardioprotective strategies intended to reduce reperfusion injury. If the phenomenon of reperfusion injury is clinically meaningful, then any intervention that can modulate the inflammatory response holds the potential to limit infarct size, to promote an improved infarct healing, and to reduce mortality.


The Immunoinflammatory Response to Ischemia and Reperfusion


When Reimer and Jennings initially described their wavefront hypothesis of AMI, little was known about the role of neutrophils, the complement cascade, and toll-like receptor mediated pathways to myocardial damage after reperfusion. Of interest, the idea of myocardial protection following myocardial infarction emerged around the same time. Since then, more than 10 distinct anti-inflammatory strategies have been tested in clinical trials to limit the adverse consequences of ischemia and reperfusion ( Table 25-2 ). With the possible exception of corticosteroids, the strategies tested in the clinic have been based on the hypothesis that the early inflammatory reaction associated to the reperfusion on an infarcted myocardium is harmful, and therefore should be down-modulated.



TABLE 25–2

Selected Randomized Clinical Trials Targeting Inflammatory and Immunity during Acute Coronary Syndrome









































































































































































































Trial, Year (n) Agent, Dose, Study Design Time of Drug to Symptoms Onset Revascularization 1° End Point Comments
Nonsteroidal anti-inflammatory drugs (NSAIDs)
TAMI-4 trial, 1989 ( n = 50) STEMI


  • Iloprost IV, 2 ng/kg/min × 48 hr



  • Open label

Fibrinolysis LVEF change at 7 days worst with iloprost The combination of rt-PA plus iloprost at the doses employed did not improve either the early or the late follow-up coronary artery patency
NUT-2, 2002 ( n = 120) UA/NSTEMI


  • Meloxicam 15 mg IV once, then 15 mg PO daily × 30 day



  • Open label

The composite of death, MI, and recurrent angina at 90 days significantly better with meloxicam No adverse complications associated with meloxicam treatment were observed. Trial performed before the widespread adoption of early invasive PCI in NSTEMI
Corticosteroids
SoluMedrol Sterile Powder AMI, 1986 ( n = 1118) STEMI


  • Methylprednisolone IV, 30 mg/kg q 3 hr × 2 vs. placebo



  • Double-blind

Within 6 hr (group 1) or 6-12 hr (group 2) Mortality at 28 days did not differ between groups Methylprednisolone not associated with myocardial rupture or cardiac aneurysm
Statins
ARMYDA-ACS, 2007, ( n = 171) NSTEMI


  • Atorvastatin PO, 80 mg vs. placebo



  • Double-blind

7 days before PCI PCI The composite of death, MI, and unplanned revascularization reduced with atorvastatin High-risk ACS patients were excluded from the study.
Adenosine and agonists
AMISTAD, 1999 ( n = 236) STEMI


  • Adenoscan IV, 70 µg/kg/min × 3 hr vs. placebo



  • Open label

Within 6 hr Fibrinolysis 99m Tc SPECT infarct size at 7 days 33% smaller with adenosine The reduction of infarct size more pronounced in patients with anterior infarction. Patients randomized to adenosine tended to experience more adverse clinical events
Marzilli et al, 2000 ( n = 54) STEMI


  • Adenosine IC, 4 mg over 1 min vs. placebo



  • Single blind

Within 3 hr Primary PCI No-reflow less frequent with adenosine. The composite of death, MI, and CHF better with adenosine The IC injection of adenosine was safe and well tolerated. No bradyarrhythmias were observed. The rates of TIMI 3 flow at the end of the PCI were significantly better in adenosine
ATTACC, 2003 ( n = 608) STEMI


  • Adenosine item IV 10 µg/kg/min × 6 hr vs. placebo



  • Double-blind

Within 6 hr Fibrinolysis Echocardiogram LVEF at hospital discharge did not differ between groups Recruitment stopped due to apparent lack of efficacy after interim analysis. Cardiovascular mortality of 8.9% with adenosine and 12.1% with placebo at 1 yr ( P = .2)
ADMIRE, 2003 ( n = 311) STEMI


  • Adenosine agonist AmP579 IV 15 vs. 30 vs. 45 µg/kg vs. placebo × 6 hr



  • Double-blind

Within 6 hr Primary PCI 99m Tc SPECT infarct size at 5-9 days did not differ between groups
AMISTAD II, 2005 ( n = 2118) STEMI


  • Adenosine IV 50 vs. 70 µg/kg/min × 3 hr vs. placebo



  • Double-blind

Within 6 hr


  • Fibrinolysis 58.3%



  • Primary PCI 40.2%

The survival free of death, CHF, and rehospitalization at 6 mo did not differ between groups Patients treated with the adenosine 70 µg/kg/min had a smaller infarct size compared with placebo (11% vs. 27% of the left ventricle, P = .02)
P-selectin inhibitors
RHAPSODY, 2003, ( n = 598) STEMI


  • rPSGL-Ig IV, 5 mg vs. 25 mg vs. 75 mg vs. placebo



  • Double-blind

Within 6 hr Fibrinolysis Time to ST-segment resolution proportional to the rPSGL-Ig doses Neither 99m Tc SPECT infarct size at 7 days nor the LVEF at 30 days differed between groups
PSALM, 2006 ( n = 88) STEMI


  • rPSGL-Ig IV, 75 vs. 150 mg vs. placebo



  • Open label

Within 6 hr Fibrinolysis 13 NH 3 myocardial blood flow and 18 FDG metabolic activity did not differ at 30 days between the groups Trial prematurely stopped after lack of efficacy shown in accompanying larger trial. No difference seen for ST-segment resolution and LVEF improvement
CD11/CD18 integrin receptor blockade
LIMIT AMI, 2001 ( n = 394) STEMI rhuMAb CD18 IV, bolus 0.5 vs. 2.0 mg/kg vs. placebo
Double-blind
Within 12 hr Fibrinolysis Corrected TIMI frame count, or the rate of TIMI 3 flow at 90 min did not differ between groups There was neither treatment effect on the infarct size nor on the rate of ECG ST-segment resolution. There was a trend toward more bacterial infections with rhuMab CD18
FESTIVAL, 2001 ( n = 60) STEMI



  • Hu23F2G IV, 0.3 vs. 1.0 mg/kg vs. placebo



  • Double-blind

Within 6 hr Primary PCI 99m Tc SPECT infarct size at 5-9 days did not differ between groups One patient found to have antibodies against Hu23F2G. No significant differences in adverse events (including infections) noted between the groups
HALT AMI, 2002 ( n = 420) STEMI


  • Hu23F2G IV, 0.3 or 1.0 mg/kg vs. placebo



  • Double-blind

Within 6 hr Primary PCI 99m Tc SPECT infarct size at 5-9 days did not differ between groups No difference in the infarct size was seen in the subgroup of patients with anterior MI or those presenting within 2 hr of symptoms onset. Clinical events at 30 days very low
Complement inhibitors
COMMA, 2003 ( n = 960) STEMI


  • C5 inhibitor Pexelizumab IV, bolus alone vs. bolus + infusion vs. placebo. B: 2 mg/kg, I: 0.05 mg/kg × 20 hr



  • Double-blind

Within 6 hr Primary PCI 72-hr CK-MB AUC did not differ between groups The mortality at 90 days significantly lower with pexelizumab bolus + infusion compared with placebo (1.8% vs. 5.9%, P = .014)
COMPLY, 2003 ( n = 943) STEMI


  • C5 inhibitor pexelizumab, same as COMMA



  • Double-blind

Within 6 hr Fibrinolysis 72-hr CK-MB AUC did not differ between groups Adjunctive pexelizumab was given 10 min after lytics on average. No significant effect on clinical outcomes
APEX-AMI, 2007 ( n = 5745) STEMI


  • C5 inhibitor pexelizumab vs. placebo, B: 2 mg/kg, I: 0.05 mg/kg x 20 hr



  • Double-blind

Within 6 hr Primary PCI Mortality at 30 days did not differ between groups The trial was intended to enroll 8500 patients initially. The composite end points of death, shock, or heart failure did not differ between groups
Thielmann, 2006 ( n = 57) STEMI C1-esterase inhibitor Berinert vs. placebo, B: 40 UI/kg IV, I: 20 UI/kg × 6 hr, Open label Within 24 hr of symptoms onset Emergency CABG surgery 24-hr TnI AUC did not differ between groups Patients treated with C1-INH within 6 hr of symptom onset had significantly lower peak maximum TnI level
ITF-1697
PARI MI, 2004 ( n = 402) STEMI


  • ITF-1697 IV, dose finding, bolus, followed by infusion (0.1, 0.5, 1.0, or 2.0 µg/kg/min) vs. placebo × 24 hr



  • Double-blind

Within 12 hr Primary PCI The HBDH AUC infarct size and clinical outcome at 30 days did not differ between groups ITF-1697 did not affect the post-PCI perfusion assessed by corrected TIMI flow, blush grade, or ST-segment resolution
Erythropoietin
Lipsic et al, 2006 ( n = 22) STEMI


  • Darbepoetin-α IV, 300 µg



  • Open-label

Within 6 hr Primary PCI Peak CK and CK-MB did not differ between groups, but were numerically higher in the darbepoetin group No adverse events were recorded during the 30-day follow-up. At 4 months, the LVEF were similar between groups (52% ± 3% for EPO vs. 48% ± 5% for placebo)
Liem et al, 2007 ( n = 51) NSTEMI


  • Epoetin-α IV, 40,000 IU vs. placebo



  • Open label

Within 8 hr of first positive troponin I 72-hr CK-MB AUC did not differ between groups The rates of reinfarction were similar at 1 yr. The authors described a blood pressure augmentation in the hours following the administration of erythropoietin
Binbrek et al, ( n = 236) STEMI


  • β-Epoetin IV, 30,000 IU vs. standard therapy



  • Open label

Within 6 hr Tenecteplase CK-MB gram equivalents (infarct size) did not differ between groups The LVEF measured by echocardiogram before discharge was similar between the groups
FX06
F.I.R.E., 2009 ( n = 234) STEMI


  • FX06 400 mg IV vs. placebo



  • Double-blind

Within 6 hr Primary PCI Late gadolinium-enhanced CMR infarct size at 5 days did not differ between groups At 5 days, FX06 was associated to a significant reduction in the necrotic core zone measured with CMR. At 40 days, this difference was no longer significant
Cyclosporine and analogues
Piot et al, 2008 ( n = 58) STEMI


  • Cyclosporine IV, 2.5 mg/kg vs. placebo



  • Single blind

Within 12 hr Primary PCI 72-hr CK AUC was significantly better in patients treated with cyclosporine, whereas the Tn I AUC was not In a substudy of 27 patients, MI size measured by MRI was 20% smaller in cyclosporine-treated patients

B, bolus; C1-INH, complement-1 inhibitors; CK, creatinine kinase; CMR, cardiac magnetic resonance; I, infusion; IC, intracoronary; IV, intravenous; ECG, electrocardiogram; FDG, fludeoxyglucose (18F); HBDH, α-hydroxybutyrate dehydrogenase; MRI, magnetic resonance imaging; PO, per os (by mouth), rt-PA; recombinant tissue-plasminogen activator, rhuMAb; recombinant human monoclonal antibody; rPSGL-Ig; recombinant P-selectin glycoprotein ligand-immunoglobulin; TnI, troponin I; UA, unstable angina.


The healing of the heart following a transmural infarct occurs in three overlapping phases: the inflammatory phase, the proliferative phase, and the maturation phase. The inflammatory phase starts when the necrosed myocytes release ligands normally sequestered away from the immune system inside the cells. Intracellular ligands, such as heat shock proteins or the fibronectin fragments, are perceived as a threat by the immune system, which activates several immunoinflammatory pathways, such as the toll-like receptor (TLR)-mediated pathways, the nuclear factor NF-κB, and the complement cascade. Of all the immune system compartments involved in the overall healing process, only a few have been shown to contribute specifically to reperfusion injury.


PMNs are a first-line defense against foreign antigens and play a key role in the acute inflammatory response following tissue injury. After a persistent coronary artery occlusion, PMNs accumulate in the myocardium, reaching a peak within 24 hours and then diminishing over the course of 1 week. In experimental models of ischemia and reperfusion, the PMN accumulation is accelerated compared with what is observed in the non-reperfused infarct. In dogs, for instance, the neutrophil count is 80% higher in the reperfused hearts compared with non-reperfused hearts. Neutrophils accumulate in the reperfused myocardium and may cause further infarction through their cytotoxic armamentarium ( Fig. 25-1 ). , When activated by appropriate stimuli, neutrophils undergo a respiratory burst that leads to the genesis of superoxide and oxygen radicals, which are major mediators of lethal cellular injury. , In addition to their direct toxic effect, oxygen radicals induce vasoconstriction and aggravate the post-ischemic coronary endothelial dysfunction. Likewise, the neutrophil-produced oxidants can depress the calcium transport in myocytes and sarcoplasmic reticulum and exacerbate ventricular dysfunction. The degranulation of activated PMNs exposes the interstitium to several proteolytic enzymes that can directly cause myocyte damage. For instance, collagenases and elastases cleave the interstitial matrix molecules into chemotactic peptide fragments, which may subsequently recruit monocytes into the necrotic myocardium. The recruitment of monocytes and other inflammatory cells is also maintained by the release of proinflammatory arachidonic acid metabolites that amplify and perpetuate the inflammatory reaction. In addition to their cytotoxic effect, neutrophils block the microcirculation to contribute to the reflow phenomenon. PMNs are larger and less compliant than red blood cells. Once activated, PMNs become less easily deformable and tend to adhere in clusters to the endothelial cells to form microcirculatory plugs. Finally, the protease and free oxygen radicals released by neutrophils can directly activate the complement cascade, that further accentuates the inflammatory reaction and the cellular damage.




FIGURE 25–1


The activation of neutrophils. Schematic diagram of activation of neutrophils, and neutrophil-derived products that participate in lethal myocardial ischemia-reperfusion injury. Reactive oxygen species generated by neutrophils or coronary vascular endothelial cells stimulate the immediate release of proinflammatory factors in ischemic-reperfused myocardium, as well as increased transcription of factors through NF-nB. These factors then activate neutrophils to generate reactive oxygen species, proteases, various cytokines and lipid mediators, and upregulate surface expression of adhesion molecules that interact with endothelium and cardiomyocytes. CD, cluster determinant; H 2 O 2 , hydrogen peroxide; Isch, ischemia; LTB 4 , leukotriene B 4 ; MIP-2, macrophage inflammatory protein-2; n-fMLP, N-formyl peptides; -O 2 , superoxide anions; -OH, hydroxyl anion; Rep, reperfusion; ROS, reactive oxygen species; XA 2 , thromboxane A 2 .

(Reproduced with permission from Vinten-Johansen J: Involvement of neutrophils in the pathogenesis of lethal myocardial reperfusion injury. Cardiovasc Res 2004;61:481-497.)


The complement system is part of the innate immune system and has a fundamental protective role against infective agents. The complement system can be activated by the classic, the alternative, and the lectin pathways, which all converge at the cleavage of C3 into C3a + C5a, two potent anaphylatoxins capable of recruiting inflammatory cells, and into C3b which flags cells for phagocytosis ( Fig. 25-2 ). Once activated, the complement system amplifies the inflammatory reaction and perpetuates an environment favorable to fight foreign invaders. In the terminal portion of the complement cascade, C5 cleavage initiates the formation of the C5b-9 membrane attack complex (MAC), which is a transmembrane channel that causes direct tissue injury through osmotic lysis. While desirable when the organism has to fight a microbiologic invader, the non-specific complement-induced amplification of the inflammatory response may be harmful to the host in the specific context of ischemia-reperfusion injury. The accumulation of complement in the ischemic heart soon after reperfusion suggests its involvement in reperfusion injury. In vivo, the inhibition of the complement cascade during myocardial infarction has been shown to result in decreased levels of proinflammatory complement byproducts (sC5b-9), , in reduced chemotaxis of polymorphonuclear leukocytes in the myocardium, and in a reduction of myocyte necrosis and apoptosis.




FIGURE 25–2


The activation of the complement system. All three activation pathways merge at the cleavage of C3 and lead to the generation of anaphylatoxins (C3a, C5a) for the recruitment of inflammatory cells, flagging of cells for phagocytosis (C3b) and the generation of the membrane attack complex (MAC: C5b-9) for cell lysis. Regulation of the complement system is extremely important and each step is controlled by several inhibitors. MASP, mannose-binding lectin-associated serine protease; MBL, mannose-binding-lectin; PAMPs, pathogen-associated molecular pattern.

(Reproduced with permission from Haahr-Pedersen S, Bjerre M, Flyvbjerg A, et al: Level of complement activity predicts cardiac dysfunction after acute myocardial infarction treated with primary percutaneous coronary intervention. J Invasive Cardiol 2009;21:13-19.)


Clinical Investigations with Anti-Inflammatory and Immunosuppressive Agents


Aspirin and the Nonsteroidal Anti-inflammatory Drugs


The interaction between platelets and the inflammatory system are numerous. It is therefore no surprise that interventions aimed at inhibiting platelet function may exert, in parallel, significant anti-inflammatory effects. Aspirin has a compelling mortality benefit during AMI. In addition to its antiplatelet properties, aspirin exerts some anti-inflammatory effects that may contribute to some of the mortality benefit observed after myocardial infarction. , In animal models, aspirin triggers the synthesis of lipoxins (known as the aspirin-triggered LXA4) that vigorously inhibits leukocyte chemotaxis during ischemia and reperfusion injury. Nonsteroidal anti-inflammatory drugs (NSAIDs), on the other hand, have been extensively studied because of their possible deleterious effect on the endothelium and on atherosclerotic plaques. In dogs, ibuprofen has been shown to reduce both the infarct size and leukocyte accumulation inside the infarcted tissue. This is in the context, however, of the well-established effect of indomethacin on impairing infarct healing and leading to aneurysm formation.


In a retrospective study meant to assess the effect of NSAIDs in the pre-aspirin era, Sajadieh and colleagues looked at the pre-randomization use of NSAIDs among the Danish Verapamil Infarction Trial (DAVIT) II trial. The authors hypothesized that NSAIDs other than aspirin may also improve the prognosis after AMI. After correction for age, sex and hypertension, the use of NSAIDs before myocardial infarction was associated with a non-significant reduction in mortality (hazard ratio [HR], 0.59; 95% confidence interval [CI], 0.28 to 1.25; P = .17), and major cardiac events (HR, 0.67; 95% CI, 0.37 to 1.19; P = .17). While this is useful for hypothesis generation, the association may well be due to confounding.


Prostacyclin (PGI 2 ) is a member of the eicosanoid family that naturally prevents the formation of clot by inhibiting platelet activity and by causing arterial vasodilatation. In experimental settings, prostacyclin was shown to facilitate fibrinolysis and reduce myocardial stunning following infarction. These early experiments served as the bases for the Thrombolysis and Angioplasty in Myocardial Infarction (TAMI)-4 trial which tested iloprost, a synthetic analogue of prostacyclin, in combination to rt-PA in 50 patients with evolving ST-segment elevation myocardial infarction (STEMI). Compared with rt-PA alone, the combination of intravenous iloprost (2 ng/kg/min for 48 hr) and rt-PA neither affected the rates of infarct-related vessel patency at 90 minutes (44% vs. 66% respectively, P = .26) nor improved the ejection fraction recovery at 7 days (+2.9% vs. −2.3% respectively, P = .05).


The inhibitors of cyclooxygenase-2 (COX-2) have been linked to increased cardiovascular risk when administered for chronic conditions. Paradoxically, a selective COX-2 inhibition has been shown to reduce the macrophage infiltration and to preserve the ventricular function after myocardial infarction in rodents. At least one trial has suggested a protective effect of short-term administration of COX-2 inhibitor to patients with acute coronary syndrome. In the Nonsteroidal Anti-Inflammatory Drugs in Unstable Angina Treatment-2 (NUT-2) pilot study, patients with non–ST-segment elevation myocardial infarction (NSTEMI) randomized to 30 days of meloxicam (a selective COX-2 inhibitor) experienced less recurrent angina, myocardial infarction, or death than the patients randomized to placebo (21.7% vs. 48.3% respectively, P = .004, n = 120) at 90 days after initial hospitalization. In this small study, the benefits were mainly driven by the high rates of recurrent angina in the placebo-treated patients. Neither the investigators nor the patients were blinded to the treatment assignment. These findings await validation in a large double-blind trial.


Corticosteroids


Corticosteroids have a pleiotropic effect on the immune system. They were among the first immunomodulators studied in AMI. Corticosteroids exert adverse effects on the cardiovascular system, such as dyslipidemia and hypertension. Since the 1970s, several case series have linked the use of corticosteroids with left ventricular free wall rupture in the days following AMI. The mechanism by which corticosteroids prevent healing of myocardial infarction is not entirely known; it may relate to impaired scar formation of the degenerative process responsible for the peripheral myopathy seen in patients treated with chronic corticosteroids. The steroid-dependent attenuation of the sympathetic innervations of the infarct region could also contribute to impaired healing. On the other hand, the powerful immunosuppressive effect of corticosteroids may modulate the inflammatory response that could have beneficial effects in the short term.


Corticosteroids have been tested in patients with acute coronary syndrome. In the Muna randomized trial, methylprednisone was compared with placebo in patients with recent-onset unstable angina. While 48 hours of methylprednisone decreased C-reactive protein levels compared with placebo, it had no apparent effect on subsequent coronary events. In fact, a trend toward a better event-free survival was seen in the placebo-control group. The trial was underpowered to address the effect of steroids on clinical events. In 1986, the Solu-Medrol Sterile Powder AMI Studies Group reported the results of a double-blind, placebo controlled trial that tested whether a single dose of methylprednisolone (30 mg/kg IV) reduced 28-day mortality in 1118 patients with recent myocardial infarction complicated by cardiac failure. Per design, the trial hypothesized that an effect of methylprednisolone could depend on time since symptom onset. When administered within 6 hours of symptoms onset, methylprednisolone did not seem to affect mortality compared with placebo (11.7% vs. 9.9%, P = NS). However, when started more than 6 hours after the onset of symptoms, Solu-Medrol significantly reduced mortality at 48 hours (10.4% vs. 14.7%, P = .04) and at 6 months ( P = .03). Interestingly, similar rates of cardiac aneurysms (1.2% vs. 0.5%, P = .45), and cardiac ruptures (1.7% vs. 2.5%, P = .47) were observed between methylprednisolone and placebo-treated patients.


In a recent meta-analysis of 11 controlled trials including 2646 patients, a 26% relative mortality reduction was observed with the use of corticosteroids within the first few hours following AMI (odds ratio [OR], 0.74; 95% CI, 0.59 to 0.94; P = .02). In a sensitivity analysis which excluded the non-randomized study, the previously observed mortality benefit with steroids was no longer present (OR, 0.95; 95% CI, 0.72 to 1.26). Importantly, no clear association was observed between myocardial rupture and corticosteroids, calling into question the conventional wisdom that steroids impair infarct healing. While none of the trials was performed in the modern era of thrombolytics and aggressive percutaneous revascularization, the available evidence suggests no evidence of either substantial benefit or harm from corticosteroids in AMI.


Statins


Just like aspirin, long-term statin administration improves the prognosis of patients across the entire spectrum of acute coronary syndromes (ACS). However, the short-term benefits of statin therapy early after ACS are equivocal. In a recent meta-analysis, the initiation of a statin within 14 days following the onset of an acute coronary event did not reduce death, myocardial infarction (MI), or stroke at 4 months. Fewer studies have assessed the protective role of statins when present before the occurrence of an ACS. In this regard, the Atorvastatin for Reduction of Myocardial Damages during Angioplasty (ARMYDA)-trial has provided interesting insights on the mechanism of action of statins. In ARMYDA, a pretreatment with 80 mg of atorvastatin at least 12 hours before the percutaneous coronary intervention (PCI) resulted in a significant reduction of the composite incidence of death, myocardial infarction, and unplanned revascularization at 30 days (5% vs. 17% for placebo, P = .01), mainly through a reduction in peri-procedural infarctions. Atorvastatin resulted in a lower average percent increase of C-reactive protein level after the PCI (63% ± 114% vs. 147% ± 274% for placebo). After adjusting for key clinical predictors (NSTEMI, left ventricular ejection fraction [LVEF] below 40%, the use of glycoprotein IIb/IIIa inhibitors and beta blockers), atorvastatin remained a significant predictor of favorable clinical outcome. In a substudy of ARMYDA, the levels of soluble intercellular cell adhesion molecule-1 (ICAM-1) and E-selectin were significantly less increased at 24 hours following the PCI in patients treated with atorvastatin, suggesting a modulatory effect of statin on leukocytes and endothelial cell interactions.


The acute benefit seen with statins in ACS patients cannot be solely explained by the cholesterol-lowering effect, which requires a longer duration of treatment. This so-called pleiotropic effect of statins could be in part due to a favorable modulation of the inflammatory system. , Animal experiments have suggested that statins can reduce the infarct size, related to a decrease in endothelial expression of P-selectin on endothelial cells and CD18 on leukocytes, leading to a reduced neutrophil extravasation into the reperfused myocardium. , Together with the stabilization of the endothelial function and the microcirculatory vasodilatation, the immunomodulatory effects of statins appear to favor survival of cardiomyocytes following ischemia/reperfusion.


Adenosine and its Agonists


Adenosine is an endogenous purine nucleoside that antagonizes several of the metabolic, biochemical and inflammatory pathways that may be involved in reperfusion injury ( Fig. 25-3 ). Adenosine and its receptor appear to be important elements of the intrinsic protective mechanism of the myocardium against ischemic insult. When subjected to hypoxia, myocardial and endothelial cells naturally release adenosine which, in turn, brings myocardial cells into a phenotype of sustained tolerance against ischemia. This phenomenon, called preconditioning, is incompletely understood but involves the inhibition of oxygen free radical formation, the repletion of cardiomyocytes and endothelial cells with high-phosphate stores, and the increase in nitric oxide bioavailability. Equally important is the cardioprotective effect of adenosine through the modulation of inflammation. Adenosine has a marked inhibitory effect on neutrophils by limiting their reactive oxygen species generation and their adherence to endothelial cells, possibly via a down-modulation of CD11/CD18 expression. In addition to its antineutrophil effects, adenosine may also serve as a metabolic switch that senses tissue injury and acts to reduce the production of inflammatory cytokines, such as tumor necrosis factor-α, interleukin-12, or macrophage inflammatory protein (MIP)-1a. In animal models of reperfusion injury, adenosine has consistently improved coronary blood flow, reduced infarct size, and improved left ventricular functional recovery.




FIGURE 25–3


Anti-inflammatory pathways impacted by adenosine. A summary of pathways of adenosine formation and catabolism in the heart and their potential modulation during ischemia-reperfusion. Broken arrows reflect regulatory processes, with plus ( + ) and minus ( − ) symbols denoting activation or inhibition of pathways during de-energization/ischemia, respectively. Whereas catabolism of adenosine to inosine, hypoxanthine, xanthine, and uric acid is shown within the myocytes, deamination also occurs extracellularly, and these reactions occur with high activity in vascular cells. AK, adenosine kinase; AMP-DA, AMP deaminase; Cr, creatine; ECTO-5′-NUC, ecto-5′-nucleotidase; HPRT, hypoxanthine phosphoribosyl transferase; 5′-NUC, 5′-nucleotidase; PCr, phosphocreatine; P i , inorganic phosphate; PKC, protein kinase C; PLC, phospholipase C; PLD, phospholipase D; PNP, purine nucleoside phosphorylase; TNF-α, tumor necrosis factor-α; XDH/XO, xanthine dehydrogenase/xanthine oxidase.

(Reproduced with permission from Headrick JP, Hack B, Ashton KJ: Acute adenosinergic cardioprotection in ischemic-reperfused hearts. Am J Physiol Heart Circ Physiol 2003;285:H1797-H1818.)


The cardioprotective effect of adenosine and its agonists has been assessed in distinct clinical scenarios, including acute coronary syndromes, high-risk PCI, and coronary artery bypass graft (CABG) surgery. The Acute Myocardial Infarction Study of ADenosine (AMISTAD)-I and AMISTAD-I trials assessed the role of adenosine in myocardial infarction. The 236-patient AMISTAD-I trial showed a 33% reduction ( P = .03) in infarct size measured by single-photon-emission computed tomography (SPECT) 99m Tc sestamibi with adenosine compared with placebo. In the subgroup of patients with anterior wall infarction, the difference was even more important, with a 67% relative reduction in final infarct size (15% vs. 45.5% of left ventricle respectively, P = .01). While the number of adverse clinical events was low, there was a numerical excess of death (10 vs. 6) and congestive heart failure (13 vs. 8) among patients treated with adenosine. These findings prompted the AMISTAD-II trial, a phase III randomized, double-blind trial designed to compare progressive doses of adenosine to placebo in 2118 patients with anterior STEMI. In this setting, adjunctive adenosine was no better than placebo at prolonging event-free survival (death, in-hospital congestive heart failure [CHF] and rehospitalization for CHF) at 6 months (84% vs. 82%, P = .43). Interestingly, in a substudy, patients treated with adenosine tended to have smaller median infarct size compared with patients treated with placebo (17% vs. 27% of the left ventricle, P = .07). Moreover, patients treated with the highest dose of adenosine (70 µg/kg/min) had the smallest infarct size, suggesting a dose-response relationship (11% of the left ventricle, P = .02 vs. placebo). As noted by the investigators, there was a significant association between the infarct size and the occurrence of CHF or death. The discrepancy between the actual effect of adenosine on infarct size and the clinical outcomes remains unexplained, but may relate to the relatively low power of this moderately sized trial. It also, however, calls into question the validity of surrogate outcomes—such as infarct size—to assess the clinical effects of investigational new drugs.


The benefits suggested by the AMISTAD trials were not confirmed by the ATTenuation by Adenosine of Cardiac Complications (ATTACC) trial. In ATTACC, 608 patients with STEMI were randomized to either adjunctive low-dose adenosine (10 µg/kg/min) or placebo, administered with fibrinolysis. The relatively small dose of adenosine was chosen after concerning rates of adverse events were observed with higher doses (40 µg/kg/min). In this context, the LVEF was similar between the adenosine and the placebo-treated patients (44% vs. 45% respectively, P = NS), along with the wall motion score index (1.53 vs. 1.50, respectively, P = NS). Based on this apparent lack of benefit, recruitment in the trial was stopped after less than two thirds of the patients originally planned had been enrolled. At 12 months, the cardiovascular mortality was 8.9% with adenosine and 12.1% with placebo (OR, 0.71; 95% CI, 0.4 to 1.2; P = .2). As was the case with AMISTAD-I, the effect size appeared to be more impressive in the subgroup of patients with anterior STEMI (OR, 0.53; 95% CI; 0.23 to 1.24; P = .09). Because of the early termination of recruitment, the results should be interpreted with caution.


In parallel to the AMISTAD and ATTACC trials, a series of trials have assessed the use of adjunctive adenosine agonist during AMI. The adenosine agonist AMP579 has mixed affinities for the adenosine A1 and A2 receptors. The A1 receptor is of major importance in mediating the anti-ischemic action of adenosine. The A2 receptor, on the other hand, has a more controversial contribution but appears to reduce the neutrophil adherence and the subsequent damages during reperfusion injury. In animal models of myocardial reperfusion injury, AMP579 has demonstrated superior cardioprotective effects compared with adenosine, possibly due to a superior inhibition of the vascular and myocardial tissue injury induced by neutrophils. The AmP579 Delivery for Myocardial Infarction REduction (ADMIRE) trial was designed to find the most efficient dose of AmP579 in patients with STEMI treated with primary PCI. To this end, 311 patients were randomized to either three different doses of AmP579 or placebo intravenously over 6 hours. No differences were seen in the final infarct size or the myocardial salvage index after adjustment for infarct-related artery, the time to reperfusion, and the initial TIMI flow. Likewise, the rates of adverse events were similar between the groups at 4 weeks and 6 months. ADMIRE did not demonstrate a benefit of AmP579 on infarct size or clinical outcomes.


Taken together, the trials presented in this section fail to show a clear clinical benefit of adjunctive adenosine with a timely reperfusion therapy. Additional studies are required, however, to fully explore the consequence of the beneficial effect of adenosine on infarct size and microvascular function.


Anti-Leukocyte Therapies


The endothelium serves as a natural barrier that protects the organ tissue from certain elements circulating in the blood. The endothelium is normally responsible to orchestrate the trafficking of leukocytes into the myocardium (see Fig. 25-3 ). In response to hypoxia, the endothelium undergoes a series of transcriptional and non-transcriptional changes similar to those seen during acute inflammation. PMNs normally migrate into the ischemic tissue. Once in place, PMNs are thought to cause direct tissue damage and to amplify the inflammatory response during the reperfusion. During an AMI, the microvascular damage and disruption of the endothelium barrier allow a massive influx of PMNs into the necrotic core and the surrounding viable myocardium. Likewise, PMNs secrete factors that disrupt the endothelial barrier and favor the formation of tissue edema.


Leukocytes transmigrate tissue through a series of complex steps that involve anchoring molecules expressed by the endothelium. Circulating neutrophils expressing P-selectin glycoprotein ligand 1 (PSGL-1) are initially caught by the endothelial receptor P-selectin (CD62P). This low-affinity interaction allows the neutrophils to slow their course and roll on the endothelium until they adhere more firmly. This process is mediated by the ligation of the leukocyte β2 integrins CD11a/CD18 and the endothelial ICAM-1. Once tightly bound to the endothelium, the leukocytes can transmigrate into the ischemic tissue, where they release toxic reactive oxygen species and other proinflammatory substances.


After a myocardial infarction, the pharmacologic interventions targeting the leukocytes can be regrouped in three distinct targets: the inhibition of leukocyte adhesion molecule synthesis (such as CD11a/CD18), the inhibition of receptor engagement and endothelial cell adhesion (such as P-selectin), and the inhibition of inflammatory mediator release, such as platelet activation factor (PAF), and leukotriene B 4 (LTB 4 ). Both P-selectin and CD11a/CD18 have recently been tested in patients with STEMI.


P-Selectin Inhibitors


P-selectin (CD62P) is an adhesion molecule that plays a critical role in the migration of leukocytes through the vascular walls. The ligand of P-selectin, PSGL-1, is constitutively expressed by leukocytes. P-selectin is involved in the adhesion of platelets to the endothelium and has been shown to play an important role in the process of atherogenesis. Convincing studies have shown that animals lacking P-selectin have a decreased tendency to develop atherosclerotic plaques. After promising animal experiments, the interference between P-selectin and PSGL-1 by the way of a monoclonal antibody was expected to facilitate the fibrinolysis and to decrease the infarct size by controlling the migration of leukocytes into the necrotic areas. Because of its anti-thrombotic and anti-inflammatory effects, P-selectin inhibition appeared to be a promising target during acute MI.


The P-selectin Antagonist Limiting Myonecrosis (PSALM) trial tested a recombinant P-selectin glycoprotein ligand-immunoglobulin (rPSGL-Ig) in patients with STEMI presenting within 6 hours of symptom onset and treated with alteplase. The engineered P-selectin antagonist combined the high-affinity ligand of the amino-terminal portion of PSGL-1 with an Fc (fragment crystallizable) region of the human IgG1. Patients were randomly assigned to a single intravenous bolus of either 75 mg of rPSGL-Ig, 150 mg of rPSGL-Ig, or placebo. The study tested the potential benefit of rPSGL-Ig on infarct size reduction using positron emission tomography to quantify the myocardial blood flow in the infarct territory, normalized for the systemic equilibrated blood pool. A total of 88 patients were enrolled before the trial was prematurely stopped by the sponsor. At 5 days, the normalized infarct size was not statistically different between the patients treated with placebo and the patients treated with either the low or the high doses of rPSGL-Ig (9.1% vs. 3.8% vs. 4.3%). The PSALM trial was stopped after a larger trial (RHAPSODY) failed to show any benefit of adjunctive rPSGL-Ig on the speed of ST-segment resolution and the reduction of infarct size in nearly 600 STEMI patients treated with fibrinolysis. , The RHAPSODY trial failed to show any significant effect of rPSGL-Ig on improvement of the LVEF, and the occurrence of death, stroke, and MI at 30 or 180 days. Unlike what was expected based on the animal data, there was a significant delay in myocardial reperfusion seen in patients treated with rPSGL-Ig, as shown by a significant prolongation of time to ST-segment resolution ( P = .008). The redundancy of the inflammatory pathways in mammals may account for the lack of efficacy of a selective inhibition of P-selectin.


CD11/CD18 Integrin Receptor Blockade


Neutrophils start to accumulate at the periphery of the necrotic myocardium as early as 8 hours after the start of the infarction. Leukocytes transmigrate after they ligate their β 2 integrins CD11a/CD18 to ICAM-1. Using a rationale similar to with the P-selectin inhibitors, the blockade CD11/CD18 integrin receptor has been tested in two independent randomized controlled trials. The LIMIT-AMI trial was designed to define the safety and the efficacy of a rhuMAb CD18, a recombinant humanized monoclonal antibody against the CD18 subunit of the β 2 integrin adhesion receptors in 394 patients with AMI. This randomized, double-blind trial compared two intravenous doses of the study drug (0.5 mg/kg vs. 2.0 mg/kg) to placebo, administered concomitantly with thrombolytic therapy. At 90 minutes after the start of fibrinolysis, the corrected TIMI frame count (primary end point) was similar between the groups, as was the percent of patients with a resolution of their ST-segment elevation resolution at 3 hours. The myocardial infarct size (assessed by 99m Tc-sestamibi SPECT) was also similar, as were the rates of adverse clinical events. Compared to placebo, the systemic administration of the study drug resulted in a peak of circulating white blood cells at 24 hours, which rapidly resolved by 72 hours, with no apparent effect on other blood compartments.


The HALT-MI trial tested whether Hu23F2G, a humanized antibody directed against all the isoforms of the CD11/CD18 integrin receptors, would reduce the infarct size in patients with AMI treated with primary PCI. The HALT-MI randomized 420 patients either to Hu23F2G (0.3 mg/kg or 1.0 mg/kg) or placebo given prior to primary PCI. The left ventricle infarct size measured by SPECT was used as the primary study end point. The study drug was administered as a single intravenous bolus over 1 to 2 minutes. The doses of Hu23F2G used in the trial were shown to saturate up to 80% of the CD11/CD18 integrin receptors for 12 to 24 hours in healthy volunteers. The final infarct size for the intention-to-treat population was similar in the 0.3 mg/kg, 1.0 mg/kg, and the placebo groups (16% vs. 17.2% vs. 16.4%, respectively; P = .80). Infarct sizes measured at baseline with the creatine kinase MB (CK-MB) area under the curve (AUC) for 24 hours were also similar. No difference in the rate of adverse clinical events at 30 days was detected. As was the case for the LIMIT-AMI trial, minor infections (mainly urinary tract infection) were more common in the active treatment groups. Together, the LIMIT-AMI and the HALT-MI trial show lack of beneficial cardiac effects of CD18 blockade in patients with AMI.


The time elapsed between the start of the myocardial infarction and the administration of the blockers of the CD11/CD18 integrin receptor varied significantly between animals and humans. This difference has been suggested as part of the explanation for the discrepant findings seen between the animal experiments and this (and other) clinical trials. In most animal experiments, the study drug was administered within 45 to 60 minutes after the ligation of the coronary artery. In the LIMIT-AMI study, the median time to symptom onset was 2.7 hours, for instance. The delays seen in humans may have resulted in a greater burden on endothelial cell barrier rupture. Assuming this were true, the greater endothelial permeability may in return have allowed a free circulation of neutrophils into the injured myocardium therefore bypassing the usual Mac-1 intercellular adhesion mechanism. Interestingly, no favorable effects were seen with the late administration of the CD11/CD18 integrin receptor blocker in experimental myocardial infarction.


Complement Inhibitors


The complement inhibitors in general and pexelizumab in particular are amongst the most studied anti-inflammatory agents in ACS. The development program for pexelizumab enrolled more than 15,000 patients with an ischemic cardiac disease in diverse randomized investigations. Pexelizumab is one of the few anti-inflammatory drugs formally tested in a phase III clinical trial of AMI.


Two inhibitors of the complement system have been tested in patients with STEMI: the classical pathway, through the inhibition of C1-(esterase); and the final common pathway, through the inhibition of C5. C1-(esterase) inhibitors have been shown to considerably reduce the reperfusion injury in translational models of AMI. , In 2002, de Zwaan and coworkers reported on the effect a C1-inhibitor purified from human plasma (Cetor; CLB, the Netherlands) and administered for 48 hours in patients with a recent AMI. The study drug was started no sooner than 6 hours after the onset of symptoms. In this series, a dose-dependent reduction of complement activity was observed. In the subgroup of patients successfully reperfused with thrombolytics, the area under the curve for troponin and creatinine kinase-MB mass was significantly reduced compared with untreated control patients (36% and 57%; P = .001). The intravenous administration of C1-(esterase) inhibitors, which prior to this experiment was limited to patients with hereditary angioedema, was deemed safe at doses up to 100 U/kg in adult patients with recent STEMI. The medication appears to have a narrow therapeutic window; extensive thrombosis was found in pigs receiving 200 U/kg of C1-inhibitor before coronary reperfusion. Safety concerns about C1-(esterase) inhibitors have been raised after 9 deaths due to venous thrombosis were observed in neonates who received more than 300 U/kg of C1-inhibitor to reduce capillary leak during major cardiac surgery. ,


More recently, another C1-(esterase) inhibitor was assessed in 57 patients with ongoing STEMI treated with reperfusion by emergency coronary artery bypass surgery. Patients were randomized to either the inhibitor (ZLB Behrig, Marburg, Germany) or matching placebo. All surgeries were performed using standard cardioplegia techniques. In this context, the peak maximum troponin I serum levels were not significantly different between the groups, showing no benefit of C1-(esterase) inhibition in this small study. A subgroup analysis suggested that the treatment could reduce the infarct size if administered within 6 hours of symptom onset. Interestingly, the efficacy of C1 inhibition was confirmed by a significant increase in the serum C1-inhibition activity along with a reduced C3c and C4 complement fragment concentration at 24 hours in the blood of patients enrolled in the active treatment arm. No drug-related adverse events were reported. Of note, no postoperative coagulation or thrombotic disorders were observed. At 30 days, the rates of death, stroke, major bleeding, and renal failure were similar between the groups.


Pexelizumab is a recombinant humanized monoclonal antibody to C5. Pexelizumab blocks the amplification of the inflammatory reaction triggered by the complement cascade by inhibiting the conversion of C5 to C5a, an anaphylatoxin, and to C5b, a precursor of the C5b-9 membrane attack complex (MAC). The COMPlement inhibition in myocardial infarction treated with thromboLYtics (COMPLY) trial was a phase II investigation designed to assess the efficacy of pexelizumab as an adjunctive to fibrinolysis in patients with ongoing STEMI. The trial randomized 943 patients to either a single pexelizumab bolus (2.0 mg/kg), or to a pexelizumab bolus plus an infusion (0.05 mg/kg/hr × 20 hr) or to matching placebo. The median infarct size, as determined by the CK-MB AUC, did not change by treatment nor did the 90-day composite incidence of death, CHF, cardiogenic shock, or stroke (bolus, 18.4%; bolus plus infusion, 19.7%; placebo, 18.6%).


In parallel to the COMPLY trial, the COMplement inhibition in Myocardial infarction treated with Angioplasty (COMMA) trial assessed whether pexelizumab decreased infarct size in STEMI patients reperfused by primary PCI. To this end, 960 patients were randomized to either a single pexelizumab bolus (2.0 mg/kg), or a pexelizumab bolus (2.0 mg/kg) with an infusion (0.05 mg/kg/hr × 20 hr) or placebo. The bolus of pexelizumab or its matching placebo was to be given before the first device activation. While the administration of pexelizumab effectively inhibited the plasma complement activity, it did not reduce the infarct size, as measured by the CK-MB AUC ( P = .89). Likewise, the 90-day composite of death, new or worsening heart failure, and stroke occurred in 8.5% of the bolus plus infusion patients versus 11.1% of the placebo patients (relative risk [RR], 0.77; 95% CI, 0.46 to 1.29). Of interest, pexelizumab given as a bolus plus an infusion was associated with a nominally significant reduction in mortality (RR, 0.30; 95% CI, 0.11 to 0.81), and in cardiogenic shock (RR, 0.55; 95% CI, 0.23 to 1.29), when compared to placebo. At 6 months, the difference in mortality was still significant (RR, 0.43; 95% CI, 0.20 to 0.94). The mortality among patients treated with a single bolus of pexelizumab was intermediate to the mortality observed with the bolus plus the infusion and the placebo groups, suggesting a dose-response relationship. The apparent contradiction between the improved survival and the lack of reduction in infarct size with pexelizumab in the COMMA trial led the investigators to hypothesize that the study drug may have mediated its effect through a reduction in inflammation at a systemic level, and/or through other effects on healing such as reduced apoptosis. In the COMMA trial, higher plasma levels of C-reactive protein and interleukin-6 (IL-6) levels at baseline, 24 hours, and 72 hours were significantly associated with increased mortality. Compared with placebo, patients treated with pexelizumab experienced a lowering of circulating levels of C-reactive protein and IL-6 at 24 hours after study drug administration (25.5 mg/L vs. 17.1 mg/L, P = .03, and 63.8 pg/mL vs. 51.0 pg/mL, P = .04, respectively). This hypothesis and the suggestion of a mortality benefit found in COMMA were then tested for validation in a phase III trial, the APEX-AMI trial.


The Assessment of Pexelizumab in Acute Myocardial Infarction (APEX AMI) trial compared pexelizumab (bolus plus infusion) to placebo as adjunctive to primary PCI. APEX-AMI was a multicenter, double-blind, placebo-controlled, phase III study that enrolled patients with anterior ST elevation or inferior elevation plus right precordial ST depression. The trial was stopped early after a large trial of pexelizumab in the setting of bypass surgery was negative. APEX AMI showed that at 30 days, there was no difference in all-cause mortality between pexelizumab and the placebo (4.1% vs. 3.9%, respectively; HR, 1.04; 95% CI, 0.80 to 1.35; P = .78). The composite end points of death, shock, or heart failure were also similar (9.0% vs. 9.2%, respectively; HR, 0.98; 95% CI, 0.83 to 1.16; P = .81). Despite the efforts to enroll high-risk patients, the surprisingly low event rate actually observed made it conceivable that the trial was underpowered to detect a treatment benefit (type II error). , However, the lack of substantial benefit of pexelizumab in patients with STEMI was recently highlighted by a meta-analysis including the COMPLY, COMMA, and APEX-AMI trials ( n = 7019). In this analysis, adding pexelizumab to mechanical or pharmacologic reperfusion did not change the rates of death (OR, 0.79; 95% CI, 0.61 to 1.03; P = .11), myocardial infarction (OR, 1.04; 95% CI, 0.89 to 1.22; P = .14), stroke (OR, 0.95; 95% CI, 0.66 to 1.38; P = .8), or congestive heart failure (OR, 1.0; 95% CI, 0.82 to 1.22; P = .99). In the same meta-analysis, pexelizumab was associated with a significant reduction in mortality when given to patients undergoing coronary artery bypass surgery (OR, 0.74; 95% CI, 0.58 to 0.94; P = .01), another condition of ischemia/reperfusion. This apparent contradiction might be explained by the variation in the timing of pexelizumab administration and/or varying role of complement in reperfusion injury in the two conditions.


ITF-1697


ITF-1697 is a chemically modified LyS-Pro tetrapeptide (Gly-(Et)Lys-Pro-Arg) that corresponds to the sequence 113 to 116 of C-reactive protein. ITF-1697 exerts an antianaphylactic activity and has been tested in at least two distinct clinical trials on patients with MI. , While virtually no preclinical information has been formally published, before the trials came out in 2004, ITF-1697 was said to reduce reperfusion injury by preventing PMN adhesion and extravasation and by limiting the increase in vascular permeability and microcapillary plugging seen during no reflow. In the Protect Against Reperfusion Injury in acute Myocardial Infarction (PARI-MI), four dose regimens of adjunctive ITF-1697 were compared with placebo in patients with AMI who were eligible for PCI. In PARI-AMI, neither a dose-relation nor a benefit could be seen in terms of infarction size, post-procedural coronary blood flow, or clinical outcome.


Emerging Anti-Inflammatory Interventions


Erythropoietin


In the past decade, our understanding of the role played by erythropoietin (EPO) has gradually shifted from a concept of strictly hematopoiesis to a broader role including anti-hypoxia. Erythropoietin appears to play an important role in the development of the heart, and its defense against injury. During embryogenesis, the inactivation of the erythropoietin receptors leads to defects in cardiac morphogenesis. The discovery that the cardiomyocytes express the receptor for erythropoietin has opened the way to a series of investigations to test for a protective role during ischemic stress. Treatment with human recombinant erythropoietin in animal models of ischemia-reperfusion has been associated with reduced myocardial infarct size and improved left ventricular functional recovery. , Erythropoietin stimulates postnatal neovascularization by enhancing endothelial precursor cell mobilization from the bone marrow. Likewise, erythropoietin inhibits cardiac myocyte hypoxia-induced apoptosis through PI3K-Akt-dependent pathways, , which could favor myocardial healing. Erythropoietin also exerts a potent anti-inflammatory effect in the myocardium, where it has been shown to directly inhibit IL-6, tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein , and to block the acute inflammatory component of reperfusion injury by inducting AP-1. Erythropoietin significantly reduces inflammatory cell infiltration and fibrosis in animal models of myocardial infarction , and chronic congestive heart failure.


It is not known whether erythropoietin has a protective effect against ischemia-reperfusion injury in humans. Recently, high endogenous erythropoietin levels were associated with a smaller infarct size in patients treated with primary percutaneous coronary intervention. The efficacy of recombinant erythropoietin is currently being tested in several small trials of patients with ACS. In a pilot study testing a single dose of epoitin-α on patients with non–ST-segment elevation ACS, no significant reduction in myocardial damage was found when compared with placebo. Instead, an increase in systolic blood pressure was observed in the hours following erythropoietin (+10 mm Hg ± 16 mm Hg for epoitin vs. −6 mm Hg ± 16 mm Hg for placebo, P = .007), which is a surprising finding for a short-term therapy. In patients with STEMI treated with primary PCI, a single intravenous high dose of darbepoetin alfa or erythropoietin was safe and well tolerated. In 2009, Binbrek and colleagues. reported on the effect of β-EPO (single 30,000 IU IV bolus before tenecteplase) compared with standard care in 236 patients admitted with STEMI within less than 6 hours of onset of symptoms. Despite EPO, the infarct size index was virtually identical in the EPO and control groups (12.4 ± 0.9 vs. 13.2 ± 0.1 creatine kinase-MB gram equivalents, respectively, data presented as mean ± SE, P = NS). At discharge, the LVEF was similar in both groups. The results of this trial prompted larger confirmatory trials that are currently underway to test the effect of EPO in patients with STEMI.


Cell Therapy


Inflammation is a putative target of cell therapy in the diseased heart. Certain cell populations may be naturally predisposed to inhibit inflammation. For instance, the mesenchymal stem cells (MSCs) have been used with apparent success in advanced inflammatory conditions, such as refractory Crohn disease or steroid-resistant graft-versus-host disease. , Mesenchymal stem cells or marrow stromal cells are a rare population in the bone marrow that forms the supportive niche required by the hematopoietic tissue to maintain their function. Transplanted MSCs have been shown to down-modulate the inflammatory response in multiple experimental models, including after AMI. MSCs secrete a vast array of pro-angiogenic cytokines and growth factors. , During in vivo experiments, MSCs transplanted into ischemic milieu deliver cytokines into the surrounding tissues, which is thought to favor vasculogenesis and reduce apoptosis. In addition to a possible regenerative effect, the paracrine secretion of cytokines and growth factors has been proposed as one of the mechanisms for the cardioprotective effects of MSCs during ischemic injury. , MSCs do not express major histocompatibility complex class II and co-stimulatory molecules. For this reason, MSCs can directly interact with cells of the immune system to modulate the anti-inflammatory environment that can promote healing and survival of damaged cells. Whether MSCs can truly escape reaction by the immune system is controversial. The clinical experience with MSCs on cardiac patients is limited, however. Compared to placebo, the intravenous administration of heterologous MSCs (Provacel) has been associates with an encouraging protective signal in patients with recent STEMI. Interestingly, a significant improvement in the forced expiratory volume in 1 second (FEV 1 ) was observed in all patients treated with cells, suggesting a possible anti-inflammatory lung effect. A larger trial use is currently underway to assess the efficacy of MSCs in patients with STEMI.


Not every cell population seems to have beneficial anti-inflammatory effect. In the ASTAMI trial, bone marrow mononuclear cells (BMMNCs) were not superior to placebo at improving the left ventricular function following a STEMI. In the study, the intracoronary injection of autologous BMMNCs resulted in a transient, yet pronounced augmentation of both the circulating IL-6 and the expression of TNF-α mRNA, along with a lesser decrease in C-reactive protein in the days following the AMI. Interestingly, investigators have suggested that the improvement in cardiac function after BMMNC therapy was associated with a transient increase in myocardial infarction. A better understanding of the role played by the immune system in cardiac repair following a myocardial infarction will open the way to better cell-based regenerative strategies.


FX06


FX06 is a naturally occurring fibrinogen product that was initially developed as a laboratory test to monitor thrombogenesis in acute coronary syndrome. FX06 is a 28-amino-acid peptide derived from the peptide sequence Bβ15-42 of human E1 fibrin fragment. , It is released by plasmin during fibrinolysis and binds to vascular endothelial (VE)-cadherin to interfere with the diapedesis of leukocytes across the endothelium. The receptor for FX06 has not been identified, but the receptor CD11c expressed by neutrophils and monocytes has been proposed. In animal models of ischemia-reperfusion, FX06 has been shown to substantially reduce the circulating levels of IL-6. Likewise, it decreased the infiltration of leukocytes into the injured myocardium, therefore reducing the infarct size and subsequent scar formation. , FX06 has a plasma half-life ranging from 11 to 17 minutes (in healthy volunteers).


In the FX06 in the Prevention of Myocardial Reperfusion Injury (F.I.R.E) trial, the interplay of fibrin fragments, leukocytes and VE-cadherin was assessed in 234 patients presenting with STEMI undergoing primary PCI within 6 hours of onset of symptoms. Patients were randomly assigned to either the study drug (two IV boluses; 200 mg before coronary guidewire crossed the lesion and 200 mg 10 minutes later) or their matching placebo. F.I.R.E used the infarct size at 5 days as a primary end point, measured by late gadolinium enhancement (LGE) on cardiac magnetic resonance (CMR), with adjustment for the rates of TIMI 0/1 flow, and the presence of collaterals before the primary PCI. Of note, the infarct size at baseline was not assessed. At 5 days, the adjusted infarct size was not different between groups (19.7% for FX06 vs. 19.8% for placebo, P = .48). Interestingly, a significant difference was seen in the size of the necrotic core zone (1.8 g vs. 4.2 g; P <.025, favoring FX06). Likewise, FX06 resulted in numerically lower rates of microvascular obstruction (27.6% vs. 37.5%, P = .09) measured by CMR. At 4 months, no significant differences in total LGE improvement could be seen by CMR. Overall, there were no differences across the groups in rates of treatment-related adverse events.


Cyclosporine and Analogues


Cyclosporine A has been used for decades as an immunosuppressive drug to prevent rejection after the transplantation of solid organs. Cyclosporine forms with cyclophilin D a complex that inhibits calcineurin, a potent transcription inducer of IL-2 and other lymphokines. More recently, cyclosporine and its analogues have been shown to inhibit the opening of mitochondrial permeability transition pores. The defective mitochondria are thought to play a major adverse role during reperfusion injury seen with organ transplantation. , The mitochondrial permeability transition pores are found in the inner membrane of the mitochondria. In normal conditions, these pores nonspecifically conduct low-molecular-weight solutes. During ischemia and reperfusion, the calcium and reactive oxygen species cause the pores to reopen, , resulting in mitochondrial depolarization with uncoupling of oxidative phosphorylation. The uncoupling of the respiratory chain causes the cells to become deplete in adenosine triphosphate (ATP), which leads to cell death. The accompanying release of pro-apoptotic cytochrome C by the mitochondria may further result in cardiomyocyte death.


The pharmacologic suppression of mitochondrial permeability transition pore during ischemia and reperfusion has recently been tested in a pilot study. Piot and colleagues tested the efficacy and safety of a single cyclosporine bolus given at the time of reperfusion in patients with STEMI treated with primary PCI. The size of the myocardial infarction as measured by the 72-hour CK AUC was significantly reduced in patients treated with cyclosporine, compared with placebo (relative infarct size reduction, 40%; P = .04). Smaller infarct sizes were also measured among cyclosporine-treated patients enrolled in the cardiac magnetic resonance sub-study (relative infarct size reduction, 27%; P = .04). The rates of adverse clinical events were similar between the groups. At 3 months, the mean left ventricular ejection fraction as measured by echocardiography were similar in both groups (cyclosporine, 50% ± 2% vs. placebo, 47% ± 3%; P = .32). In the study, the blood concentration of cyclosporine was nearly undetectable in a majority of patients 12 hours after the administration of the drug.


In addition to its effect on the mitochondrial permeability transition pores, cyclosporine has additional effects that could contribute to myocardial protection during ischemia/reperfusion. In vitro, cyclosporine A appears to be effective at lowering the neutrophil chemotaxis, as well as release of toxic lysozyme and superoxide anion in response to stimulation. Likewise, cyclosporine regulates the expression of inducible nitric oxide synthase (iNOS), which has been shown to protect the myocardium in ischemic conditions. Proof-of-concept clinical trials with N -methyl-4-isoleucine cyclosporin (N1M811C), a cyclosporine analogue with no known immunosuppressive effect, will help to clarify the role played by the mitochondrial permeability transition pore during the reperfusion injury.


p38 Mitogen-Activated Protein Kinase Inhibitor


The p38 mitogen-activated protein kinase (MAPK) plays a key role in the initiation and the amplification of inflammatory processes. P38 MAPK activity increases early after myocardial infarction and appears to stay activated for several weeks thereafter. In vitro, p38 MAPK has been involved in the regulation of myocyte hypertrophy and apoptosis. In experimental settings, p38 MAPK inhibition has resulted in infarct size reduction in the acute phase. Likewise, it has been shown to favorably remodel the left ventricle in the chronic phase following an acute infarction. Importantly, p38 MAPK inhibition has modified the atherogenic process in animal models of chronic coronary artery disease. Because of its simultaneous action on the coagulation system and the inflammatory system, p38 MAPK is an appealing target for the treatment of acute coronary syndromes.


In humans, p38 MAPK has been shown to upregulate coagulation processes by increasing the expression of tissue factor on the endothelial cells and the monocytes. , In a phase I trial, oral inhibitors of the p38 MAPK attenuated the surge of circulating proinflammatory cytokines TNF-α, IL-6, and IL-8 in response to endotoxin stimulation. , Oral p38 MAPK inhibitors have been tested with limited success in chronic inflammatory conditions, such as rheumatoid arthritis and Crohn disease. , Clinical trials are under way to investigate p38 MAPK as a target for cardiovascular protection in patients with ACS.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jan 22, 2019 | Posted by in CARDIOLOGY | Comments Off on Inflammation and Immunity as Targets for Drug Therapy in Acute Coronary Syndrome

Full access? Get Clinical Tree

Get Clinical Tree app for offline access