Abstract

Atherosclerosis is a chronic inflammatory disease. Pathophysiological similarities between chronic infections and atherosclerosis triggered interest in a clinical association between these conditions. Various infectious microbes have been linked to atherosclerotic vascular disease in epidemiological studies. However, this association failed to satisfy the Koch’s postulates of causation with multiple clinical trials demonstrating inefficacy of anti-infective therapies in mitigating atherosclerotic cardiovascular events. Identification of underlying pathophysiological mechanisms and experience with vaccination against various infectious agents has ushered a new avenue of efforts in the development of an anti-atherosclerotic vaccine. Studies in animal models have identified various innate and adaptive immune pathways in atherosclerosis. In this review, we discuss the patho-biological link between chronic infections and atherosclerosis, evaluate existing evidence of animal and human trials on the association between infections and cardiovascular disease and introduce the concept of an anti-atherosclerotic vaccine.

Introduction

Atherosclerosis is now considered a chronic inflammatory disease. Various stimuli that trigger and sustain this state of heightened inflammation have been identified. Chronic infections represent one such stimulus that share the common pathophysiological milieu of chronic inflammation. Various bacteria and viruses have been shown to have a direct effect on the vascular endothelium as well as an indirect effect by systemic cytokine release, both of which contribute to accelerated atherosclerosis. Infectious agents that have been linked to atherosclerotic disease include, but not limited to Chlamydia pneumoniae, Porphyromonas gingivalis, Helicobacter pylori, influenza A virus, hepatitis C virus, cytomegalovirus (CMV), and human immunodeficiency virus (HIV). However, there are significant differences in the strength of the data supporting their association. Emerging evidence supporting the role of immunity in atherosclerosis and the discovery of molecular mimicry between oxidized low density lipoprotein cholesterol (ox-LDL) and bacterial cell wall polysaccharide has opened new avenues in the search for an anti-atherosclerotic vaccine. This has led to interest in targeting infective agents in an effort to mitigate the atherosclerotic process. Initial data from animal studies has fuelled multiple human clinical trials of anti-infective therapies to alleviate atherosclerosis.

In this review, we discuss the patho-biological link between chronic infections and atherosclerosis, evaluate existing evidence of animal and human trials on the association between infections and cardiovascular disease and highlight key features concerning the development of an anti-atherosclerotic vaccine.

Pathobiologic mechanisms of atherosclerosis development

Infections have long been implicated in the initiation, progression, and rupture (plaque instability and thrombosis) of atherosclerotic plaque. The ‘response to injury’ hypothesis conceptualizes atherosclerosis as a pathway of chronic inflammation and lipid accumulation in response to endothelial injury.1 Although sero-epidemiologic studies demonstrating an association between infectious agents and clinical atherosclerosis are conflicting, one of the strongest arguments in favour of the ‘infection hypothesis’ is based on the common substrate of chronic inflammation.2 Many studies propose the ‘direct’ (invasion of cells causing accelerated plaque growth through local effects) and ‘indirect’ mechanisms (systemic expression of inflammatory cytokines which catalyze development of plaque) by which infectious agents contribute to atherogenesis. Here, we review some of the key mechanisms implicated in the ‘infection hypotheses’ of atherosclerosis.

Local effects

Numerous studies have shown the presence of bacterial and viral microorganisms in the atherosclerotic plaque. Pathogens may be harboured in a latent state or replicate in cells such as macrophages creating a chronic inflammatory environment. Most implicated organisms are intracellular microbes which exert their effects from within the cell, evading the body’s immune mechanisms.3  ,  4 Major local pathogenic effects of various microbes are listed in Table 1.

Table 1

Summary of local pro atherosclerotic mechanisms in chronic infections

Phase of atherogenesisPathogenPathogenic effect
Lesion formation  
  • Activation of endothelium

  • Migration of leucocytes

  • Formation of lipid core

  • CMV, Cp, Pg, HIV, HCV, Influenza

  • Cp, Pg, CMV, HIV, Influenza

  • Cp, CMV

  • HIV, Pg

  • ↑ Expression of adhesion molecules

  • ↑ Production of MCP-1

  • Promotes oxidation of LDL

  • ↑ Expression of scavenger receptors→ ↑ uptake of ox-LDL

  • Promote influx of foam cells

Lesion progression  
  • Proliferation of SMC

  • Cp, CMV

  • Pg, CMV

  • Resistance to apoptosis

  • ↑ production of growth factors

Lesion instability  
  • Rupture

  • Thrombosis

  • Cp, Hp

  • Cp, HIV, CMV

  • Cp, CMV, HSV

  • Altered vasomotor tone

  • ↑ production of MMPs

  • Anticoagulant → Pro-coagulant milieu

Phase of atherogenesisPathogenPathogenic effect
Lesion formation  
  • Activation of endothelium

  • Migration of leucocytes

  • Formation of lipid core

  • CMV, Cp, Pg, HIV, HCV, Influenza

  • Cp, Pg, CMV, HIV, Influenza

  • Cp, CMV

  • HIV, Pg

  • ↑ Expression of adhesion molecules

  • ↑ Production of MCP-1

  • Promotes oxidation of LDL

  • ↑ Expression of scavenger receptors→ ↑ uptake of ox-LDL

  • Promote influx of foam cells

Lesion progression  
  • Proliferation of SMC

  • Cp, CMV

  • Pg, CMV

  • Resistance to apoptosis

  • ↑ production of growth factors

Lesion instability  
  • Rupture

  • Thrombosis

  • Cp, Hp

  • Cp, HIV, CMV

  • Cp, CMV, HSV

  • Altered vasomotor tone

  • ↑ production of MMPs

  • Anticoagulant → Pro-coagulant milieu

Summary of postulated local effects of infectious agents in atherogenesis.

CMV, cytomegalovirus; Cp, Chlamydia pneumoniae; Pg, Porphyromonas gingivalis; HIV, human immunodeficiency virus; HCV, hepatitis C virus; Hp, Helicobacter pylori; HSV, herpes simplex virus; MCP-1, macrophage chemoattractant protein-1; LDL, low density lipoprotein; ox-LDL, oxidized LDL; MMPs, matrix metalloproteinases.

Table 1

Summary of local pro atherosclerotic mechanisms in chronic infections

Phase of atherogenesisPathogenPathogenic effect
Lesion formation  
  • Activation of endothelium

  • Migration of leucocytes

  • Formation of lipid core

  • CMV, Cp, Pg, HIV, HCV, Influenza

  • Cp, Pg, CMV, HIV, Influenza

  • Cp, CMV

  • HIV, Pg

  • ↑ Expression of adhesion molecules

  • ↑ Production of MCP-1

  • Promotes oxidation of LDL

  • ↑ Expression of scavenger receptors→ ↑ uptake of ox-LDL

  • Promote influx of foam cells

Lesion progression  
  • Proliferation of SMC

  • Cp, CMV

  • Pg, CMV

  • Resistance to apoptosis

  • ↑ production of growth factors

Lesion instability  
  • Rupture

  • Thrombosis

  • Cp, Hp

  • Cp, HIV, CMV

  • Cp, CMV, HSV

  • Altered vasomotor tone

  • ↑ production of MMPs

  • Anticoagulant → Pro-coagulant milieu

Phase of atherogenesisPathogenPathogenic effect
Lesion formation  
  • Activation of endothelium

  • Migration of leucocytes

  • Formation of lipid core

  • CMV, Cp, Pg, HIV, HCV, Influenza

  • Cp, Pg, CMV, HIV, Influenza

  • Cp, CMV

  • HIV, Pg

  • ↑ Expression of adhesion molecules

  • ↑ Production of MCP-1

  • Promotes oxidation of LDL

  • ↑ Expression of scavenger receptors→ ↑ uptake of ox-LDL

  • Promote influx of foam cells

Lesion progression  
  • Proliferation of SMC

  • Cp, CMV

  • Pg, CMV

  • Resistance to apoptosis

  • ↑ production of growth factors

Lesion instability  
  • Rupture

  • Thrombosis

  • Cp, Hp

  • Cp, HIV, CMV

  • Cp, CMV, HSV

  • Altered vasomotor tone

  • ↑ production of MMPs

  • Anticoagulant → Pro-coagulant milieu

Summary of postulated local effects of infectious agents in atherogenesis.

CMV, cytomegalovirus; Cp, Chlamydia pneumoniae; Pg, Porphyromonas gingivalis; HIV, human immunodeficiency virus; HCV, hepatitis C virus; Hp, Helicobacter pylori; HSV, herpes simplex virus; MCP-1, macrophage chemoattractant protein-1; LDL, low density lipoprotein; ox-LDL, oxidized LDL; MMPs, matrix metalloproteinases.

Lesion formation: activation of endothelium, migration of leucocytes, and formation of lipid core

Insult to the endothelial cell monolayer by shear stress, excess of ox-LDL or hyperglycaemia results in production of reactive oxygen species (ROS), chemoattractant cytokines and induction of adhesion molecules which enable entry and migration of inflammatory cells into the sub-intimal space. Multiple infectious agents, including C. pneumonia, CMV, periodontal pathogens, HIV, HCV, Influenza, have been shown to induce expression of adhesion molecules on endothelial cells.5  ,  6 Heat shock protein 60 (HSP 60) isolated from P. gingivalis has been shown to induce a toll-like receptor (TLR) 4 mediated increase in Intercelluar adhesion molecule-1 (ICAM-1), Vascular cell adhesion molecule-1 (VCAM-1), and ox-LDL receptor (LOX-1) expression in mouse vessels.7 An increased production of monocyte chemoattractant protein-1 (MCP-1), has been found to be induced by many pathogens, including C. pneumoniae, periodontal organisms, CMV, HIV, and Influenza A virus.8  ,  9

Lipid accumulation in the sub-endothelial layers occurs by internalization of ox-LDL, largely mediated by specialized receptors such as lectin-like ox-LDL receptor-1 (LOX-1), scavenger receptors (SR)-AI/II, SRBI, CD36 and TLR.10  Chlamydia pneumoniae has been shown to bind to and activate TLRs in vitro and upregulate the expression of LOX-1 in vivo.11 Increase in ox-LDL in turn increases the production of E- selectin, I-CAM-1, VCAM-1 in endothelial cells. Cytomegalovirus has been shown to mediate this through increased mRNA expression of class A SRs and expression of CD-36.12 HIV-1 Nef protein has been demonstrated to impair ATP-binding cassette transporter A1-dependent cholesterol efflux from human macrophages, and promote formation of foam cells.13 HIV-1 virus is also known to promote transendothelial migration as well as inhibit reverse transendothelial migration. This is further enhanced by increased levels of TNF-α in HIV+ serum.14 Lipopolysaccharide from P. gingivalis also increases expression of granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-1b, IL-10, and IL-12 in both macrophages and foam cells.

Lesion progression: migration and proliferation of smooth muscle cells and other key cells

Various infections have been associated with increased smooth muscle cell (SMC) proliferation in the vessel wall. Cytomegalovirus has been shown to increase SMC proliferation and inhibit apoptosis through suppression of tumour suppressor gene p53.15 In addition, CMV infection leads to enhanced levels of several growth factors including transforming growth factor β1, fibroblast growth factors, and epidermal growth factors which contribute to endothelial proliferation. Chlamydia pneumoniae infected cells have also been shown to be resistant to apoptosis.16 Gingipain, a cysteine protease of P. gingivalis has been shown to increase proliferation of aortic SMCs while another recent study showed it to increase angiopoetin 2 expression within the vessel layers and stimulate migration of SMCs.17  ,  18 Together, these changes might be expected to lead to accumulation of infected SMCs in the vessel wall and contribute to plaque growth.

Lesion rupture: thrombosis and plaque instability

Impaired vasomotor tone .i.e. decreased vasodilator and increased constrictor activity is believed to contribute to acute coronary events. Liuba et al  .  19 demonstrated in mice and swine infected with C. pneumoniae alone or coinfected with H. pylori that these infections alter vasomotor tone mediated through nitric oxide.20 Another mechanism that has been long recognized is the switch from anticoagulant to procoagulant milieu seen in human vascular endothelial cells infected with C. pneumoniae, CMV, and HSV. Vercellotti et al.21 reported that HSV infection of endothelium resulted in a marked increase in thrombin-induced platelet adhesion with a concomitant decrease in prostacyclin secretion in response to thrombin, while CMV infected endothelial cells may exhibit a procoagulant response via activation of pro-inflammatory cascade.22 Monocytes infected with CMV and C. pneumoniae have enhanced clotting mediated via increased generation of tissue factor and plasminogen activator inhibitor (PAI-1) secretion.23 Influenza virus infection also increases PAI-1 levels and decreases endothelial nitric oxide synthase expression. Matrix metalloproteinases (MMPs) have been implicated in thinning of fibrous cap and plaque destabilization. C. pneumoniae, through upregulation of LOX-1, increases levels of MMP-2 and MMP-9. While the glycoprotein 120 component of HIV-1 has been shown to increase levels of MMP-2 and MMP-9 in brain microvessel cells, the Tat protein has been shown to upregulate MMP-9 in monocytes.24 We have previously reported that patients with HCV have increased acute coronary events compared with HCV negative patients, despite similar angiographic coronary disease burden.25 Similar results have been reported in HIV patients as well.26 Enhanced plaque instability resulting from infection induced increase in MMP activity could partly explain these clinical findings. These studies implicate pathways by which incidence of vascular events is increased in patients with chronic infections.

Systemic effects

Potentiation of systemic inflammation

Regardless of pathogens found in situ in the plaque, infections induce systemic inflammation via release of cytokines and activate the immune system, both innate (TLR, HSP, and inflammasome signalling) as well as the adaptive arm (Th1, Th17 activation). Interferon alpha, which is produced in response to viral infections, is a potent inflammatory amplifier in the context of atheromatous plaque. Studies have shown that Hepatitis C viral proteins initiate a pro-inflammatory state by promoting an imbalanced T helper (Th)1/Th2 cytokine ratio.27 Other viruses, such as CMV and influenza, have also been shown to increase the plasma levels of interferon-γ, ILs and TNF-α and enhance monocyte differentiation.28 Recent studies show that periodontal pathogens such as P. gingivalis and Actinomycetes comitans promote TH17 subset responses both within the spleen and within the atherosclerotic lesions, which in turn increases the release of a host of potent cytokines such as IL-1β, IL-6, and IL-17.29  ,  30

Inflammasomes are multi-protein cytosolic complexes that integrate several pathogen-triggered signalling cascades ultimately leading to caspase-1 activation and generation of pro-inflammatory cytokines including interleukin IL-18 and IL-1β. Recently, P. gingivalis was found to increase the aortic induction of NLRP3 inflammasome receptor gene.31 HIV-1 as well as HCV have been found to activate inflammasomes in human monocytes resulting in the above mentioned effects.32  C. pnemoniae and Influenza have been shown to decrease the anti -inflammatory properties of HDL, contributing to indirect progression.33

Molecular mimicry and role of heat shock proteins

There is increasing evidence for the role of molecular mimicry in modulating immune responses to infectious agents in atherosclerosis. Protein components of microbial agents ‘mimic’ host proteins, and mount an immune response directed against host proteins which are homologous to the pathogenic antigens. Heat shock proteins have also been implicated in this process. Heat shock proteins are proteins that are phylogenetically highly conserved in mammalian and bacterial species, and the shared homology lead to extensive immunological cross-reactivity. Augmented expression of HSPs occurs on the cell surface in times of stress including infection and inflammation. Human atherosclerotic plaques express a variety of inflammatory cytokines, most of which can enhance HSP expression.34 Serum antibodies to HSP60 have been seen to cross-react with those of human HSP, C. pneumoniae HSP, Escherichia. coli HSP (GroEL), and that of H. pylori.35 Cross reactivity between anti-CMV antibodies and HSP60 has also been recently described.

Antibodies against bacterial HSP can hence induce and exacerbate the established inflammatory milieu thereby promoting plaque progression and instability.36 Plasma levels of anti-HSP60 antibodies have been correlated with atherosclerosis burden and occurrence of acute coronary syndromes.37 Kol et al.38 demonstrated that Chlamydial HSP60 mediates the activation of endothelial cells and SMCs, and induce TNF-α and MMP production by macrophages in the atherosclerotic plaques. Chlamydial HSP60 also was demonstrated to promote a Th1 immune response via maturation of monocyte derived dendritic cells.39

Thus, various chronic infections have been implicated in their association with atherosclerosis, both by epidemiological and pathophysiological association. Local and systemic effects of infections mimic and perhaps contribute to various steps in the atherosclerotic process (Figure 1).

Figure 1

Schematic of various pro-atherosclerotic pathways activated in chronic infection. The blue arrows denote direct effects of the microorganism on vascular endothelium and the orange arrows depict systemic effects that are pro-atherosclerotic.

Anti-infective therapy and atherosclerosis

To test the hypothesis that eradicating infectious stimuli would diminish the chronic inflammatory response and hence reduce cardiovascular events, several trials of anti-infective therapy have been conducted in animal models of atherosclerotic disease as well as in humans.

Animal studies

The vast majority of anti-infective studies have focused on infection with C. pneumonia, which is ubiquitous and often subclinical, so a course of targeted antibiotics could theoretically treat an infection in a large population. In addition, macrolide antibiotics which target C. pneumonia are thought to have anti-inflammatory properties, and could theoretically contribute to plaque stability. Muhlestein et al.  40 inoculated rabbits with C. pneumoniae (or placebo) and treated these animals with azithromycin, and evaluated aortic atherosclerosis and intimal thickening. They found that C. pneumoniae infection accelerated intimal wall thickening and the extent of atherosclerosis; importantly, treatment with azithromycin decreased the extent of atherosclerosis. These data were contradicted by Rothstein et al.  41 who reported no effect of azithromycin on aortic lesion size in mice infected with C. pneumoniae even though aortic lesion size was larger compared with placebo prior to antibiotic therapy.42 Madan et al.  43 studied the effect of treatment with doxycycline in P. gingivalis-associated atherosclerosis in an ApoE heterozygote murine model. They found that doxycycline decreased circulating pro-inflammatory cytokine levels as well as atherosclerotic lesion development. Others evaluated the effect of metronidazole treatment prior to P. gingivalis inoculation and demonstrated a significant decrease in circulating inflammatory markers and atherosclerotic lesion size in mice that were pretreated.44 Ayada et al  .  45 studied the effect of H. pylori infection in mice. These investigators were careful to only inoculate via the GI tract, in order to replicate the natural route of infection, as there are questions as to the amount of systemic inflammation that H. pylori can induce when administered by parenteral route. Helicobacter pylori infection was associated with increased size and thickness of aortic lesions, compared with non-infected controls. These authors then extrapolated their results to show that treatment with triple drug therapy (lasonoprazole, amoxicillin, clarithromycin) could decrease the atherogenic effects of gastrointestinal H. pylori infection. These findings from animal studies have fuelled interest in studying the effect of anti-infective therapies in humans.

Human studies

The possible link between anti-infective therapy and development or progression of atherosclerosis in animal studies led to several human studies to investigate the possible connection between anti-infective therapy and development of atherosclerosis in humans. The results of some of the major trials are summarized in Table 2. As evident, most human trials of anti-infective therapies have not shown any significant cardio-protection. However, all trials were heterogeneous in treatment choices, duration and cardiovascular end points. Initial human trials used short treatment courses of macrolide antibiotics. The largest trials of antibiotic therapy for atherosclerosis were PROVE IT TIMI (Pravastatin or Atorvastatin Evaluation and Infection Therapy - Thrombolysis in Myocardial Infarction) and ACES (Azithromycin and Coronary Events Study), and enrolled more than 4,000 patients each. The ACES trial used azithromycin, while PROVE IT-TIMI trial used gatifloxacin. There was no difference in end-points of coronary heart disease events, or cardiac death in either of these trials.

Table 2

Summary of antibiotic interventions in human clinical trials of atherosclerosis46–55

StudySample sizeAntibioticTreatment durationFollow-up durationClinical settingOutcome
In support of anti-infective therapy preventing atherosclerosis
CLARIFY148Clarithromycin3 months18 monthsACS (Non-Q)Reduced risk of ischaemic CV events
Against anti-infective therapy preventing atherosclerosis
ACADEMIC302Azithromycin3 months2 yearsCHDNo reduction in CV events
ACES4012Azithromycin12 months4 yearsCHDNo reduction in cardiac events
ANTIBIO872Roxithromycin6 weeks1 yearAMINo reduction in cardiac events
AZACS1439Azithromycin5 days6 monthsACSNo reduction in ischaemic CV events
ISAR-31020Roxithromycin1 month6–12 monthsPost-PCINo prevention of re-stenosis in patients with low C. pneumoniae titers;Reduced risk of restenosis in patients with high Chlamydia pneumoniae titers
PROVE IT TIMI4162Gatifloxacin18 months2 yearsACSNo reduction in cardiac events
ROXIS202Roxithromycin30 days6 monthsACS (Non-Q)Reduced risk of death and reinfarction
WIZARD7724Azithromycin3 months3 yearsCHDNo reduction in cardiac events
StudySample sizeAntibioticTreatment durationFollow-up durationClinical settingOutcome
In support of anti-infective therapy preventing atherosclerosis
CLARIFY148Clarithromycin3 months18 monthsACS (Non-Q)Reduced risk of ischaemic CV events
Against anti-infective therapy preventing atherosclerosis
ACADEMIC302Azithromycin3 months2 yearsCHDNo reduction in CV events
ACES4012Azithromycin12 months4 yearsCHDNo reduction in cardiac events
ANTIBIO872Roxithromycin6 weeks1 yearAMINo reduction in cardiac events
AZACS1439Azithromycin5 days6 monthsACSNo reduction in ischaemic CV events
ISAR-31020Roxithromycin1 month6–12 monthsPost-PCINo prevention of re-stenosis in patients with low C. pneumoniae titers;Reduced risk of restenosis in patients with high Chlamydia pneumoniae titers
PROVE IT TIMI4162Gatifloxacin18 months2 yearsACSNo reduction in cardiac events
ROXIS202Roxithromycin30 days6 monthsACS (Non-Q)Reduced risk of death and reinfarction
WIZARD7724Azithromycin3 months3 yearsCHDNo reduction in cardiac events

ACS, acute coronary syndrome; CHD, coronary heart disease; AMI, acute myocardial infarction; PCI, percutaneous coronary intervention.

Table 2

Summary of antibiotic interventions in human clinical trials of atherosclerosis46–55

StudySample sizeAntibioticTreatment durationFollow-up durationClinical settingOutcome
In support of anti-infective therapy preventing atherosclerosis
CLARIFY148Clarithromycin3 months18 monthsACS (Non-Q)Reduced risk of ischaemic CV events
Against anti-infective therapy preventing atherosclerosis
ACADEMIC302Azithromycin3 months2 yearsCHDNo reduction in CV events
ACES4012Azithromycin12 months4 yearsCHDNo reduction in cardiac events
ANTIBIO872Roxithromycin6 weeks1 yearAMINo reduction in cardiac events
AZACS1439Azithromycin5 days6 monthsACSNo reduction in ischaemic CV events
ISAR-31020Roxithromycin1 month6–12 monthsPost-PCINo prevention of re-stenosis in patients with low C. pneumoniae titers;Reduced risk of restenosis in patients with high Chlamydia pneumoniae titers
PROVE IT TIMI4162Gatifloxacin18 months2 yearsACSNo reduction in cardiac events
ROXIS202Roxithromycin30 days6 monthsACS (Non-Q)Reduced risk of death and reinfarction
WIZARD7724Azithromycin3 months3 yearsCHDNo reduction in cardiac events
StudySample sizeAntibioticTreatment durationFollow-up durationClinical settingOutcome
In support of anti-infective therapy preventing atherosclerosis
CLARIFY148Clarithromycin3 months18 monthsACS (Non-Q)Reduced risk of ischaemic CV events
Against anti-infective therapy preventing atherosclerosis
ACADEMIC302Azithromycin3 months2 yearsCHDNo reduction in CV events
ACES4012Azithromycin12 months4 yearsCHDNo reduction in cardiac events
ANTIBIO872Roxithromycin6 weeks1 yearAMINo reduction in cardiac events
AZACS1439Azithromycin5 days6 monthsACSNo reduction in ischaemic CV events
ISAR-31020Roxithromycin1 month6–12 monthsPost-PCINo prevention of re-stenosis in patients with low C. pneumoniae titers;Reduced risk of restenosis in patients with high Chlamydia pneumoniae titers
PROVE IT TIMI4162Gatifloxacin18 months2 yearsACSNo reduction in cardiac events
ROXIS202Roxithromycin30 days6 monthsACS (Non-Q)Reduced risk of death and reinfarction
WIZARD7724Azithromycin3 months3 yearsCHDNo reduction in cardiac events

ACS, acute coronary syndrome; CHD, coronary heart disease; AMI, acute myocardial infarction; PCI, percutaneous coronary intervention.

Among viral infections, HCV and HIV have been established to have a significant association with atherosclerotic heart disease. Recent advances in anti-viral therapies have led to the possibility of a complete cure in HCV. Whether achieving a sustained virological response to anti-HCV therapy leads to a reduction in atherosclerotic cardiovascular end points is an area of active research. In the era of interferon-based therapy, studies have shown a reduction in incidence of ischaemic stroke [hazard ratio (HR) 0.62] and acute coronary events (HR 0.77) in patients who achieved a sustained virological response over an 8-year follow up.56  ,  57 Impact of newer anti-viral regimens which have a much higher virological clearance rate, on cardiovascular end points is yet to be explored. In HIV patients, effect of anti-retroviral therapy on cardiovascular outcomes has remained controversial. In the randomized Strategies for Management of Antiretroviral Therapy trial, interruption of anti-retroviral therapy was associated with increased levels of pro inflammatory cytokines and incident myocardial infarction, suggesting that viral suppression might lead to decreased inflammatory signalling and reduced adverse cardiac events.58 However, a later large prospective study of more than 20 000 HIV patients on anti-retroviral therapy showed that the risk of incident myocardial infarction was significantly higher in patients who received protease inhibitors, even after adjusting for traditional cardiac risk factors.59 This elevated cardiac risk was attributed to protease inhibitor induced metabolic dysregulation. Whether, protease inhibitors currently approved for HCV therapy have similar cardiovascular risk profile is yet to be determined.

Overall, in most clinical trials of anti-infective agents, antibiotic choices, treatment regimens and therapeutic duration were adequate but remained ineffective. Negative results of these trials highlight the paucity in our understanding of the complex biological interactions in the pathobiology of atherosclerosis. Pathogens are perhaps not integral culprits in the progression of atherosclerosis, but rather co-exist and are incidentally noted in the atherosclerotic plaques. If bacterial pathogens were integral components, one would have seen a clear and strong correlation with anti-infective therapy slowing the progression of disease. However, several arguments explaining the relative inefficacy of antibiotic trials in reducing atherosclerotic endpoints can be justified. First, presence of persistent low grade infection that is not responsive to antibiotic therapy could explain persistent low grade inflammation. Secondly, end points in most the clinical trials were cardiovascular endpoints in patients with established vascular disease, which may or may not be modified by antibiotic therapy. Thirdly, the issue of antibiotic resistance with chronic treatment cannot be properly addressed. In summary, albeit animal experiments and a few small human clinical trials showed promise in the role of antibiotic therapy, larger studies have generally been negative.

Vaccination and atherosclerosis

As discussed above, various infections have been linked to atherosclerosis. This ‘infection hypothesis’ lends itself to possible alteration of the atherosclerotic process through treatment or prevention of infections. Given this interaction between infections and atherosclerosis, several investigators have looked at the effects of vaccinations against these organisms on the atherosclerotic process.60 In animal models, exposure to P. gingivalis accelerated atherosclerosis while immunization against it induced attenuation of atherosclerosis.61 Koizumi et al  .  62 employed nasal immunization using the outer membrane protein of P. gingivalis and demonstrated a significant reduction of atherosclerotic lesion size in the aortic sinuses of mice, along with a decrease in concentration of circulating inflammatory cytokines. Streptococcus pneumoniae is another bacterium that has been linked to atherosclerosis, perhaps driven by its molecular mimicry with oxLDL.63 Immunization against S. pneumoniae results in the production of protective anti-phosphorylcholine (PC) antibodies. These possibly result in cross-reactivity with ox-LDL, as suggested by the presence of a naturally found IgM that recognizes epitopes in both ox-LDL and the PC head groups on the surface of apoptotic cells, thus inhibiting their uptake by macrophages.64 In one study, when homozygous LDL-receptor knockout (LDL  /  ) mice were actively immunized against S. pneumoniae, it resulted in increased ox-LDL-specific antibodies and reduced atherosclerosis.65 Another group used a PC conjugate to immunize Apolipoprotein E homozygous knockout (ApoE−/−) mice. Similar to the previous study, this resulted in an increase in both anti-PC and ox-LDL antibodies, with reduced atherosclerosis.

Further elucidation of immunological mechanisms of atherosclerosis (Figure 2) has fuelled interest in vaccination as a potential preventive strategy. Encouraging results in animal studies have led to clinical evaluation of vaccines as a means of reducing atherosclerotic cardiac events. In initial single centre studies, pneumococcal vaccination was associated with a decrease of more than 50% in the rate myocardial infarction at two year follow up.66 However, data from the large prospective California Mens Health study of 84,170 participants failed to reveal a beneficial impact of pneumococcal vaccination on outcomes of acute stroke or myocardial infarction.67 The most exciting effect that an established routine vaccine has had on cardiovascular outcomes is the effect of the influenza vaccine. An increased rate of death and myocardial infarction (MI) have been noted during the influenza season.68 While the mechanism of action is unknown, several studies suggest that influenza vaccination leads to reduction in cardiovascular events such as ischaemia, rehospitalizations after MI, and cardiovascular death.69–71 A meta-analysis provided further support for the use of influenza vaccination in the secondary prevention of cardiovascular events.72 The cardiovascular effect of this vaccine could be related to the effect of antibody mediated inactivation of the influenza virus, which has been shown to directly invade vascular tissue and accelerate atherosclerosis by locally induced inflammation. There is continued mounting evidence in favour of annual influenza vaccination leading to reduced cardiovascular morbidity, stroke, and all-cause mortality in patients with known cardiovascular disease; in addition, there is no evidence of negative effects of the vaccine in the recipients.73  ,  74 The American Heart Association and the American College of Cardiology now advocate its use for secondary prevention in individuals with known cardiovascular disease (Class I, Level B).75

Figure 2

Schematic depicting various immune pathways triggered by infections. Modulations of these pathways with vaccination hold promise. Red arrows denote atherogenic pathways, green arrows denote natural anti-atherogenic pathways, and the broken blue arrows denote pathways that can to be modulated by immunization. (Treg—regulator T cells).

Anti-atherosclerosis vaccine

Given the complex interactions between immune response (innate and adaptive) and atherogenesis, modulation of this immune response towards an athero-protective path makes intuitive sense in the battle against atherosclerosis. One of the biggest challenges in this regard has been the identification of key antigens which when targeted will favourably alter the atherosclerotic process.76 To that effect, many studies have been conducted and others are underway. Unlike vaccinations against the exogenous antigens found on microbes described earlier, numerous studies exploring vaccinations against endogenous antigens have been performed. The most commonly studied antigens have included native or modified homologous LDL since LDL has the strongest causative link with atherosclerosis. Immunization of experimental animals with homologous native or modified LDL (oxidized or MDA modified) has generally resulted in a reduction in atherosclerosis. As a major component of LDL, ApoB100 was an attractive initial target for the development of an anti-atherosclerotic vaccine and has fuelled significant research in this area which is beyond the scope of the current review. We would like to refer the interested reader to recently published literature on this topic elsewhere.60  ,  77

Summary

There is a strong epidemiological association between infections and cardiovascular disease. Chronic infections and atherosclerosis share multiple biological mechanisms with ample experimental evidence supporting the pro-atherosclerotic nature of various bacterial and viral infections (Central Illustration). Despite pathophysiological correlation, anti-infective therapy has failed to satisfy Koch’s postulate of a causative link with multiple human trials demonstrating negative results. Identifying the appropriate target pathway has been the Achilles heel is in our efforts to mitigate the atherosclerotic response. Is it the microorganism per se or the myriad of signalling pathways that are activated in chronic infection that are pro-atherogenic? With negative clinical trial results and increasing data on adverse effects of prolonged antibiotic therapy on both cardiac and non-cardiac outcomes, enthusiasm towards designing more anti-infective clinical trials is dwindling. In this conjecture, development of an atherosclerosis vaccine that targets various signalling pathways related to infections may hold promise.

Central Illustration

Schematic of the pathogenic mechanisms induced by chronic infections and role of anti-atherosclerosis vaccination (IL, Interleukins, IFN, Interferon, TNF, tumour necrosis factor, TLRs, toll like receptors, MMP, matrix metalloproteinase, HSP, heat shock protein, Th, T helper cells, PAI, Plasminogen activator inhibitor, GM-CSF, granulocyte monocyte colony stimulating factor).

Conflict of interest: none declared.

References

1

Libby
 
P
,
Ridker
 
PM
,
Hansson
 
GK
;
Leducq Transatlantic Network on Atherothrombosis
.
Inflammation in atherosclerosis: from pathophysiology to practice
.
J Am Coll Cardiol
 
2009
;
54
:
2129
2138
.

2

Pant
 
S
,
Deshmukh
 
A
,
Gurumurthy
 
GS
,
Pothineni
 
NV
,
Watts
 
TE
,
Romeo
 
F
,
Mehta
 
JL
.
Inflammation and atherosclerosis–revisited
.
J Cardiovasc Pharmacol Ther
 
2014
;
19
:
170
178
.

3

Campbell
 
LA
,
Rosenfeld
 
ME.
 
Infection and atherosclerosis development
.
Arch Med Res
 
2015
;
46
:
339
350
.

4

Rosenfeld
 
ME.
 
Inflammation and atherosclerosis: direct versus indirect mechanisms
.
Curr Opin Pharmacol
 
2013
;
13
:
154
160
.

5

Adinolfi
 
LE
,
Restivo
 
L
,
Zampino
 
R
,
Guerrera
 
B
,
Lonardo
 
A
,
Ruggiero
 
L
,
Riello
 
F
,
Loria
 
P
,
Florio
 
A.
 
Chronic HCV infection is a risk of atherosclerosis. Role of HCV and HCV-related steatosis
.
Atherosclerosis
 
2012
;
221
:
496
502
.

6

Haidari
 
M
,
Wyde
 
PR
,
Litovsky
 
S
,
Vela
 
D
,
Ali
 
M
,
Casscells
 
SW
,
Madjid
 
M.
 
Influenza virus directly infects, inflames, and resides in the arteries of atherosclerotic and normal mice
.
Atherosclerosis
 
2010
;
208
:
90
96
.

7

Huang
 
CY
,
Shih
 
CM
,
Tsao
 
NW
,
Lin
 
YW
,
Shih
 
CC
,
Chiang
 
KH
,
Shyue
 
SK
,
Chang
 
YJ
,
Hsieh
 
CK
,
Lin
 
FY.
 
The GroEL protein of Porphyromonas gingivalis regulates atherogenic phenomena in endothelial cells mediated by upregulating toll-like receptor 4 expression
.
Am J Transl Res
 
2016
;
8
:
384
404
.

8

Lei
 
L
,
Li
 
H
,
Yan
 
F
,
Li
 
Y
,
Xiao
 
Y.
 
Porphyromonas gingivalis lipopolysaccharide alters atherosclerotic-related gene expression in oxidized low-density-lipoprotein-induced macrophages and foam cells
.
J Periodontal Res
 
2011
;
46
:
427
437
.

9

Park
 
IW
,
Wang
 
JF
,
Groopman
 
JE.
 
HIV-1 Tat promotes monocyte chemoattractant protein-1 secretion followed by transmigration of monocytes
.
Blood
 
2001
;
97
:
352
358
.

10

Li
 
D
,
Mehta
 
JL.
 
Antisense to LOX-1 inhibits oxidized LDL-mediated upregulation of monocyte chemoattractant protein-1 and monocyte adhesion to human coronary artery endothelial cells
.
Circulation
 
2000
;
101
:
2889
2895
.

11

Campbell
 
LA
,
Lee
 
AW
,
Rosenfeld
 
ME
,
Kuo
 
CC.
 
Chlamydia pneumoniae induces expression of pro-atherogenic factors through activation of the lectin-like oxidized LDL receptor-1
.
Pathog Dis
 
2013
;
69
:
7195
7197
.

12

Carlquist
 
JF
,
Muhlestein
 
JB
,
Horne
 
BD
,
Hart
 
NI
,
Lim
 
T
,
Habashi
 
J
,
Anderson
 
JG
,
Anderson
 
JL.
 
Cytomegalovirus stimulated mRNA accumulation and cell surface expression of the oxidized LDL scavenger receptor, CD36
.
Atherosclerosis
 
2004
;
177
:
53
59
.

13

Mujawar
 
Z
,
Rose
 
H
,
Morrow
 
MP
,
Pushkarsky
 
T
,
Dubrovsky
 
L
,
Mukhamedova
 
N
,
Fu
 
Y
,
Dart
 
A
,
Orenstein
 
JM
,
Bobryshev
 
YV
,
Bukrinsky
 
M
,
Sviridov
 
D.
 
Human immunodeficiency virus impairs reverse cholesterol transport from macrophages
.
PLoS Biol
 
2006
;
4
:
e365.

14

Maisa
 
A
,
Hearps
 
AC
,
Angelovich
 
TA
,
Pereira
 
CF
,
Zhou
 
J
,
Shi
 
MD
,
Palmer
 
CS
,
Muller
 
WA
,
Crowe
 
SM
,
Jaworowski
 
A.
 
Monocytes from HIV-infected individuals show impaired cholesterol efflux and increased foam cell formation after transendothelial migration
.
AIDS
 
2015
;
29
:
1445
1457
.

15

Tanaka
 
K
,
Zou
 
JP
,
Takeda
 
K
,
Ferrans
 
VJ
,
Sandford
 
GR
,
Johnson
 
TM
,
Finkel
 
T
,
Epstein
 
SE.
 
Effects of human cytomegalovirus immediate-early proteins on p53-mediated apoptosis in coronary artery smooth muscle cells
.
Circulation
 
1999
;
99
:
1656
1659
.

16

Paland
 
N
,
Rajalingam
 
K
,
Machuy
 
N
,
Szczepek
 
A
,
Wehrl
 
W
,
Rudel
 
T.
 
NF-kappaB and inhibitor of apoptosis proteins are required for apoptosis resistance of epithelial cells persistently infected with Chlamydophila pneumoniae
.
Cell Microbiol
 
2006
;
8
:
1643
1655
.

17

Cao
 
C
,
Ji
 
X
,
Luo
 
X
,
Zhong
 
L.
 
Gingipains from Porphyromonas gingivalis promote the transformation and proliferation of vascular smooth muscle cell phenotypes
.
Int J Clin Exp Med
 
2015
;
8
:
18327
18334
.

18

Zhang
 
B
,
Khalaf
 
H
,
Sirsjö
 
A
,
Bengtsson
 
T.
 
Gingipains from the periodontal pathogen Porphyromonas gingivalis play a significant role in regulation of angiopoietin 1 and angiopoietin 2 in human aortic smooth muscle cells
.
Infect Immun
 
2015
;
83
:
4256
4265
.

19

Liuba
 
P
,
Pesonen
 
E
,
Paakkari
 
I
,
Batra
 
S
,
Forslid
 
A
,
Kovanen
 
P
,
Pentikäinen
 
M
,
Persson
 
K
,
Sandström
 
S.
 
Acute Chlamydia pneumoniae infection causes coronary endothelial dysfunction in pigs
.
Atherosclerosis
 
2003
;
167
:
215
222
.

20

Liuba
 
P
,
Karnani
 
P
,
Pesonen
 
E
,
Paakkari
 
I
,
Forslid
 
A
,
Johansson
 
L
,
Persson
 
K
,
Wadström
 
T
,
Laurini
 
R.
 
Endothelial dysfunction after repeated Chlamydia pneumoniae infection in apolipoprotein E-knockout mice
.
Circulation
 
2000
;
102
:
1039
1044
.

21

Vercellotti
 
GM.
 
Proinflammatory and procoagulant effects of herpes simplex infection on human endothelium
.
Blood Cells
 
1990
;
16
:
209
215
.

22

van Dam-Mieras
 
MC
,
Muller
 
AD
,
van Hinsbergh
 
VW
,
Mullers
 
WJ
,
Bomans
 
PH
,
Bruggeman
 
CA.
 
The procoagulant response of cytomegalovirus infected endothelial cells
.
Thromb Haemost
 
1992
;
68
:
364
370
.

23

Bouwman
 
JJM
,
Visseren
 
FLJ
,
Bosch
 
MC
,
Bouter
 
KP
,
Diepersloot
 
RJA.
 
Procoagulant and inflammatory response of virus-infected monocytes
.
Eur J Clin Invest
 
2002
;
32
:
759
766
.

24

Louboutin
 
J-P
,
Agrawal
 
L
,
Reyes
 
BAS
,
Van Bockstaele
 
EJ
,
Strayer
 
DS.
 
HIV-1 gp120-induced injury to the blood-brain barrier: role of metalloproteinases 2 and 9 and relationship to oxidative stress
.
J Neuropathol Exp Neurol
 
2010
;
69
:
801
816
.

25

Pothineni
 
NV
,
Rochlani
 
Y
,
Vallurupalli
 
S
,
Kovelamudi
 
S
,
Ahmed
 
Z
,
Hakeem
 
A
,
Mehta
 
JL.
 
Comparison of angiographic burden of coronary artery disease in patients with versus without hepatitis C infection
.
Am J Cardiol
 
2015
;
116
:
1041
1044
.

26

Theodoropoulos
 
K
,
Mennuni
 
MG
,
Sartori
 
S
,
Meelu
 
OA
,
Yu
 
J
,
Baber
 
U
,
Stefanini
 
GG
,
Mastoris
 
I
,
Moreno
 
P
,
Dangas
 
GD
,
Mehran
 
R
,
Sharma
 
SK
,
Kini
 
AS.
 
Quantitative angiographic characterisation of coronary artery disease in patients with human immunodeficiency virus (HIV) infection undergoing percutaneous coronary intervention
.
EuroIntervention
 
2017
;
12
:
1757
1765
.

27

Niessner
 
A
,
Shin
 
MS
,
Pryshchep
 
O
,
Goronzy
 
JJ
,
Chaikof
 
EL
,
Weyand
 
CM.
 
Synergistic proinflammatory effects of the antiviral cytokine interferon-alpha and Toll-like receptor 4 ligands in the atherosclerotic plaque
.
Circulation
 
2007
;
116
:
2043
2052
.

28

Cassol
 
E
,
Cassetta
 
L
,
Alfano
 
M
,
Poli
 
G.
 
Macrophage polarization and HIV-1 infection
.
J Leukoc Biol
 
2010
;
87
:
599
608
.

29

Vliegen
 
I
,
Duijvestijn
 
A
,
Grauls
 
G
,
Herngreen
 
S
,
Bruggeman
 
C
,
Stassen
 
F.
 
Cytomegalovirus infection aggravates atherogenesis in apoE knockout mice by both local and systemic immune activation
.
Microbes Infect
 
2004
;
6
:
17
24
.

30

Cai
 
Y
,
Kobayashi
 
R
,
Hashizume-Takizawa
 
TK-OT.
 
Porphyromonas gingivalis infection enhances Th17 responses for development of atherosclerosis
.
Arch Oral Biol
 
2014
;
59
:
1183
1191
.

31

Yamaguchi
 
Y
,
Kurita-Ochiai
 
T
,
Kobayashi
 
R
,
Suzuki
 
T
,
Ando
 
T.
 
Activation of the NLRP3 inflammasome in Porphyromonas gingivalis-accelerated atherosclerosis
.
Pathog Dis
 
2015
;
73
.

32

Chattergoon
 
MA
,
Latanich
 
R
,
Quinn
 
J
,
Winter
 
ME
,
Buckheit
 
RW
,
Blankson
 
JN
,
Pardoll
 
D
,
AL.
 
HIV and HCV activate the inflammasome in monocytes and macrophages via endosomal Toll-like receptors without induction of type 1 interferon
.
PLoS Pathog
 
2014
;
10
:
e1004082.

33

Guo
 
H
,
Gao
 
J
,
Taxman
 
DJ
,
Ting
 
JPY
,
Su
 
L.
 
HIV-1 infection induces interleukin-1β production via TLR8 protein-dependent and NLRP3 inflammasome mechanisms in human monocytes
.
J Biol Chem
 
2014
;
289
:
21716
21726
.

34

Pockley
 
AG.
 
Heat shock proteins, inflammation, and cardiovascular disease
.
Circulation
 
2002
;
105
:
1012
1017
.

35

Mayr
 
M
,
Kiechl
 
S
,
Willeit
 
J
,
Wick
 
G
,
Xu
 
Q.
 
Infections, immunity, and atherosclerosis: associations of antibodies to Chlamydia pneumoniae, Helicobacter pylori, and cytomegalovirus with immune reactions to heat-shock protein 60 and carotid or femoral atherosclerosis
.
Circulation
 
2000
;
102
:
833
839
.

36

Schett
 
G
,
Xu
 
Q
,
Amberger
 
A
,
Van der Zee
 
R
,
Recheis
 
H
,
Willeit
 
J
,
Wick
 
G.
 
Autoantibodies against heat shock protein 60 mediate endothelial cytotoxicity
.
J Clin Invest
 
1995
;
96
:
2569
2577
.

37

Xu
 
Q
,
Schett
 
G
,
Perschinka
 
H
,
Mayr
 
M
,
Egger
 
G
,
Oberhollenzer
 
F
,
Willeit
 
J
,
Kiechl
 
S
,
Wick
 
G.
 
Serum soluble heat shock protein 60 is elevated in subjects with atherosclerosis in a general population
.
Circulation
 
2000
;
102
:
14
20
.

38

Kol
 
A
,
Sukhova
 
GK
,
Lichtman
 
AH
,
Libby
 
P.
 
Chlamydial heat shock protein 60 localizes in human atheroma and regulates macrophage tumor necrosis factor-alpha and matrix metalloproteinase expression
.
Circulation
 
1998
;
98
:
300
307
.

39

Ausiello
 
CM
,
Fedele
 
G
,
Palazzo
 
R
,
Spensieri
 
F
,
Ciervo
 
A
,
Cassone
 
A.
 
60-kDa heat shock protein of Chlamydia pneumoniae promotes a T helper type 1 immune response through IL-12/IL-23 production in monocyte-derived dendritic cells
.
Microbes Infect
 
2006
;
8
:
714
720
.

40

Muhlestein
 
JB
,
Anderson
 
JL
,
Hammond
 
EH
,
Zhao
 
L
,
Trehan
 
S
,
Schwobe
 
EP
,
Carlquist
 
JF.
 
Infection with Chlamydia pneumoniae accelerates the development of atherosclerosis and treatment with azithromycin prevents it in a rabbit model
.
Circulation
 
1998
;
97
:
633
636
.

41

Rothstein
 
NM
,
Quinn
 
TC
,
Madico
 
G
,
Gaydos
 
CA
,
Lowenstein
 
CJ.
 
Effect of azithromycin on murine arteriosclerosis exacerbated by Chlamydia pneumoniae
.
J Infect Dis
 
2001
;
183
:
232
238
.

42

Blessing
 
E
,
Campbell
 
LA
,
Rosenfeld
 
ME
,
Chesebro
 
B
,
Kuo
 
CC.
 
A 6 week course of azithromycin treatment has no beneficial effect on atherosclerotic lesion development in apolipoprotein E-deficient mice chronically infected with Chlamydia pneumoniae
.
J Antimicrob Chemother
 
2005
;
55
:
1037
1040
.

43

Madan
 
M
,
Bishayi
 
B
,
Hoge
 
M
,
Messas
 
E
,
Amar
 
S.
 
Doxycycline affects diet- and bacteria-associated atherosclerosis in an ApoE heterozygote murine model: cytokine profiling implications
.
Atherosclerosis
 
2007
;
190
:
62
72
.

44

Amar
 
S
,
Wu
 
SC
,
Madan
 
M.
 
Is Porphyromonas gingivalis cell invasion required for atherogenesis? Pharmacotherapeutic implications
.
J Immunol
 
2009
;
182
:
1584
1592
.

45

Ayada
 
K
,
Yokota
 
K
,
Hirai
 
K
,
Fujimoto
 
K
,
Kobayashi
 
K
,
Ogawa
 
H
,
Hatanaka
 
K
,
Hirohata
 
S
,
Yoshino
 
T
,
Shoenfeld
 
Y
,
Matsuura
 
E
,
Oguma
 
K.
 
Regulation of cellular immunity prevents Helicobacter pylori-induced atherosclerosis
.
Lupus
 
2009
;
18
:
1154
1168
.

46

Gurfinkel
 
E
,
Bozovich
 
G
,
Beck
 
E
,
Testa
 
E
,
Livellara
 
B
,
Mautner
 
B.
 
Treatment with the antibiotic roxithromycin in patients with acute non-Q-wave coronary syndromes: the final report of the ROXIS study
.
Eur Heart J
 
1999
;
20
:
121
127
.

47

Anderson
 
JL
,
Muhlestein
 
JB
,
Carlquist
 
J
,
Allen
 
A
,
Trehan
 
S
,
Nielson
 
C
,
Hall
 
S
,
Brady
 
J
,
Egger
 
M
,
Horne
 
B
,
Lim
 
T.
 
Randomized secondary prevention trial of azithromycin in patients with coronary artery disease and serological evidence for Chlamydia pneumoniae infection: the azithromycin in coronary artery disease: elimination of myocardial infection with chlamydia (ACADEMIC) study
.
Circulation
 
1999
;
99
:
1540
1547
.

48

Neumann
 
F
,
Kastrati
 
A
,
Miethke
 
T
,
Pogatsa-Murray
 
G
,
Mehilli
 
J
,
Valina
 
C
,
Jogethaei
 
N
,
da Costa
 
CP
,
Wagner
 
H
,
Schömig
 
A.
 
Treatment of Chlamydia pneumoniae infection with roxithromycin and effect on neointima proliferation after coronary stent placement (ISAR-3): a randomised, double-blind, placebo-controlled trial
.
Lancet
 
2001
;
357
:
2085
2089
.

49

Cercek
 
B
,
Shah
 
PK
,
Noc
 
M
,
Zahger
 
D
,
Zeymer
 
U
,
Matetzky
 
S
,
Maurer
 
G
,
Mahrer
 
P
;
AZACS Investigators
.
Effect of short-term treatment with azithromycin on recurrent ischaemic events in patients with acute coronary syndrome in the Azithromycin in Acute Coronary Syndrome (AZACS) trial: a randomised controlled trial
.
Lancet
 
2003
;
361
:
809
813
.

50

Burkhardt
 
U
,
Zahn
 
R
,
Höffler
 
U
,
Siegler
 
KE
,
Frilling
 
B
,
Weber
 
M
,
Gottwik
 
M
,
Wehr
 
M
,
Seidel
 
F
,
Rosocha
 
S
,
Tebbe
 
U
,
Senges
 
J.
 
Antibody levels against Chlamydia pneumoniae and outcome of roxithromycin therapy in patients with acute myocardial infarction: results from a sub-study of the randomised Antibiotic Therapy in Acute Myocardial Infarction (ANTIBIO) trial
.
Z Kardiol
 
2004
;
93
:
671
678
.

51

Sinisalo
 
J
,
Mattila
 
K
,
Valtonen
 
V,
,
Anttonen
 
O,
,
Juvonen
 
J,
,
Melin
 
J,
,
Vuorinen-Markkola
 
H,
,
Nieminen
 
MS
;
Clarithromycin in Acute Coronary Syndrome Patients in Finland (CLARIFY) Study Group
.
Effect of 3 months of antimicrobial treatment with clarithromycin in acute non-q-wave coronary syndrome
.
Circulation
 
2002
;
105
:
1555
1560
.

52

Sinisalo
 
J
,
Mattila
 
K
,
Valtonen
 
V
,
Anttonen
 
O
,
Juvonen
 
J
,
Melin
 
J
,
Vuorinen-Markkola
 
H
,
Nieminen
 
MS
;
Clarithromycin in Acute Coronary Syndrome Patients in Finland (CLARIFY) Study Group
.
Azithromycin for the secondary prevention of coronary heart disease events: the WIZARD study: a randomized controlled trial
.
JAMA
 
2003
;
290
:
1459
1466
.

53

Cannon
 
CP,
,
Braunwald
 
E,
,
McCabe
 
CH,
,
Grayston
 
JT
,
Muhlestein
 
B,
,
Giugliano
 
RP,
,
Cairns
 
R
,
Skene
 
AM
;
Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Myocardial Infarction 22 Investigators
.
Antibiotic treatment of Chlamydia pneumoniae after acute coronary syndrome
.
N Engl J Med
 
2005
;
352
:
1646
1654
.

54

Grayston
 
JT
,
Kronmal
 
RA
,
Jackson
 
LA
,
Parisi
 
AF
,
Muhlestein
 
JB
,
Cohen
 
JD
,
Rogers
 
WJ
,
Crouse
 
JR
,
Borrowdale
 
SL
,
Schron
 
E
,
Knirsch
 
C
;
ACES Investigators
.
Azithromycin for the secondary prevention of coronary events
.
N Engl J Med
 
2005
;
352
:
1637
1645
.

55

Stone
 
AF
,
Mendall
 
MA
,
Kaski
 
JC
,
Edger
 
TM
,
Risley
 
P
,
Poloniecki
 
J
,
Camm
 
AJ
,
Northfield
 
TC.
 
Effect of treatment for Chlamydia pneumoniae and Helicobacter pylori on markers of inflammation and cardiac events in patients with acute coronary syndromes: South Thames Trial of Antibiotics in Myocardial Infarction and Unstable Angina (STAMINA)
.
Circulation
 
2002
;
106
:
1219
1223
.

56

Hsu
 
YC
,
Lin
 
JT
,
Ho
 
HJ
,
Kao
 
YH
,
Huang
 
YT
,
Hsiao
 
NW
,
Wu
 
MS
,
Liu
 
YY
,
Wu
 
CY.
 
Antiviral treatment for hepatitis C virus infection is associated with improved renal and cardiovascular outcomes in diabetic patients
.
Hepatology
 
2014
;
59
:
1293
1302
.

57

Hsu
 
YC
,
Ho
 
HJ
,
Huang
 
YT
,
Wang
 
HH
,
Wu
 
MS
,
Lin
 
JT
,
Wu
 
CY.
 
Association between antiviral treatment and extrahepatic outcomes in patients with hepatitis C virus infection
.
Gut
 
2015
;
64
:
495
503
.

58

Strategies for Management of Antiretroviral Therapy (SMART) Study Group
,
El-Sadr
 
WM
,
Lundgren
 
J
,
Neaton
 
JD
,
Gordin
 
F
,
Abrams
 
D
,
Arduino
 
RC
,
Babiker
 
A
,
Burman
 
W
,
Clumeck
 
N
,
Cohen
 
CJ
,
Cohn
 
D
,
Cooper
 
D
,
Darbyshire
 
J
,
Emery
 
S
,
Fätkenheuer
 
G
,
Gazzard
 
B
,
Grund
 
B
,
Hoy
 
J
,
Klingman
 
K
,
Losso
 
M
,
Markowitz
 
N
,
Neuhaus
 
J
,
Phillips
 
A
,
Rappoport
 
C.
 
CD4+ count-guided interruption of antiretroviral treatment
.
N Engl J Med
 
2006
;
355
:
2283
2296
.

59

Data Collection on Adverse Events of Anti-HIV drugs (D:A:D) Study Group
,
Smith
 
C
,
Sabin
 
CA
,
Lundgren
 
JD
,
Thiebaut
 
R
,
Weber
 
R
,
Law
 
M
,
Monforte
 
Ad
,
Kirk
 
O
,
Friis-Moller
 
N
,
Phillips
 
A
,
Reiss
 
P
,
El Sadr
 
W
,
Pradier
 
C
,
Worm
 
SW.
 
Factors associated with specific causes of death amongst HIV-positive individuals in the D:A:D Study
.
AIDS
 
2010
;
24
:
1537
1548
.

60

Shah
 
PK
,
Chyu
 
KY
,
Dimayuga
 
PC
,
Nilsson
 
J.
 
Vaccine for atherosclerosis
.
J Am Coll Cardiol
 
2014
;
64
:
2779
2791
.

61

Miyamoto
 
T
,
Yumoto
 
H
,
Takahashi
 
Y
,
Davey
 
M
,
Gibson
 
FC
 3rd
,
Genco
 
CA.
 
Pathogen-accelerated atherosclerosis occurs early after exposure and can be prevented via immunization
.
Infect Immun
 
2006
;
74
:
1376
1380
.

62

Koizumi
 
Y
,
Kurita-Ochiai
 
T
,
Oguchi
 
S
,
Yamamoto
 
M.
 
Nasal immunization with porphyromonas gingivalis outer membrane protein decreases P. gingivalis-induced atherosclerosis and inflammation in spontaneously hyperlipidemic mice
.
Infect Immun
 
2008
;
76
:
2958
2965
.

63

Caligiuri
 
G
,
Khallou-Laschet
 
J
,
Vandaele
 
M
,
Gaston
 
AT
,
Delignat
 
S
,
Mandet
 
C
,
Kohler
 
HV
,
Kaveri
 
SV
,
Nicoletti
 
A.
 
Phosphorylcholine-targeting immunization reduces atherosclerosis
.
J Am Coll Cardiol
 
2007
;
50
:
540
546
.

64

Frostegård
 
J.
 
Low level natural antibodies against phosphorylcholine: a novel risk marker and potential mechanism in atherosclerosis and cardiovascular disease
.
Clin Immunol
 
2010
;
134
:
47
54
.

65

Binder
 
CJ
,
Hörkkö
 
S
,
Dewan
 
A
,
Chang
 
MK
,
Kieu
 
EP
,
Goodyear
 
CS
,
Shaw
 
PX
,
Palinski
 
W
,
Witztum
 
JL
,
Silverman
 
GJ.
 
Pneumococcal vaccination decreases atherosclerotic lesion formation: molecular mimicry between streptococcus pneumoniae and oxidized LDL
.
Nat Med
 
2003
;
9
:
736
743
.

66

Lamontagne
 
F
,
Garant
 
MP
,
Carvalho
 
JC
,
Lanthier
 
L
,
Smieja
 
M
,
Pilon
 
D.
 
Pneumococcal vaccination and risk of myocardial infarction
.
CMAJ
 
2008
;
179
:
773
777
.

67

Tseng
 
HF
,
Slezak
 
JM
,
Quinn
 
VP
,
Sy
 
LS
,
Van den Eeden
 
SK
,
Jacobsen
 
SJ.
 
Pneumococcal vaccination and risk of acute myocardial infarction and stroke in men
.
Jama
 
2010
;
303
:
1699
1706
.

68

Hebsur
 
S
,
Vakil
 
E
,
Oetgen
 
WJ
,
Kumar
 
PN
,
Lazarous
 
DF.
 
Influenza and coronary artery disease: exploring a clinical association with myocardial infarction and analyzing the utility of vaccination in prevention of myocardial infarction
.
Rev Cardiovasc Med
 
2014
;
15
:
168
175
.

69

Ciszewski
 
A
,
Bilinska
 
ZT
,
Brydak
 
LB
,
Kepka
 
C
,
Kruk
 
M
,
Romanowska
 
M
,
Ksiezycka
 
E
,
Przyluski
 
J
,
Piotrowski
 
W
,
Maczynska
 
R
,
Ruzyllo
 
W.
 
Influenza vaccination in secondary prevention from coronary ischaemic events in coronary artery disease: FLUCAD study
.
Eur Heart J
 
2008
;
29
:
1350
1358
.

70

Gurfinkel
 
EP
,
de la Fuente
 
RL
,
Mendiz
 
O
,
Mautner
 
B.
 
Influenza vaccine pilot study in acute coronary syndromes and planned percutaneous coronary interventions: the FLU vaccination acute coronary syndromes (FLUVACS) study
.
Circulation
 
2002
;
105
:
2143
2147
.

71

Phrommintikul
 
A
,
Kuanprasert
 
S
,
Wongcharoen
 
W
,
Kanjanavanit
 
R
,
Chaiwarith
 
R
,
Sukonthasarn
 
A.
 
Influenza vaccination reduces cardiovascular events in patients with acute coronary syndrome
.
Eur Heart J
 
2011
;
32
:
1730
1735
.

72

Udell
 
JA
,
Zawi
 
R
,
Bhatt
 
DL
,
Keshtkar-Jahromi
 
M
,
Gaughran
 
F
,
Phrommintikul
 
A
,
Ciszewski
 
A
,
Vakili
 
H
,
Hoffman
 
EB
,
Farkouh
 
ME
,
Cannon
 
CP.
 
Association between influenza vaccination and cardiovascular outcomes in high-risk patients: a meta-analysis
.
JAMA
 
2013
;
310
:
1711
1720
.

73

Siscovick
 
DS
,
Raghunathan
 
TE
,
Lin
 
D
,
Weinmann
 
S
,
Arbogast
 
P
,
Lemaitre
 
RN
,
Psaty
 
BM
,
Alexander
 
R
,
Cobb
 
LA.
 
Influenza vaccination and the risk of primary cardiac arrest
.
Am J Epidemiol
 
2000
;
152
:
674
677
.

74

Smeeth
 
L
,
Thomas
 
SL
,
Hall
 
AJ
,
Hubbard
 
R
,
Farrington
 
P
,
Vallance
 
P.
 
Risk of myocardial infarction and stroke after acute infection or vaccination
.
N Engl J Med
 
2004
;
351
:
2611
2618
.

75

AHA; ACC; National Heart, Lung, and Blood Institute
,
Smith
 
SC
 Jr
,
Allen
 
J
,
Blair
 
SN
,
Bonow
 
RO
,
Brass
 
LM
,
Fonarow
 
GC
,
Grundy
 
SM
,
Hiratzka
 
L
,
Jones
 
D
,
Krumholz
 
HM
,
Mosca
 
L
,
Pearson
 
T
,
Pfeffer
 
MA
,
Taubert
 
KA.
 
AHA/ACC guidelines for secondary prevention for patients with coronary and other atherosclerotic vascular disease: 2006 update endorsed by the national heart, lung, and blood institute
.
J Am Coll Cardiol
 
2006
;
47
:
2130
2139
.

76

Palinski
 
W
,
Miller
 
E
,
Witztum
 
JL.
 
Immunization of low density lipoprotein (LDL) receptor-deficient rabbits with homologous malondialdehyde-modified LDL reduces atherogenesis
.
Proc Natl Acad Sci USA
 
1995
;
92
:
821
825
.

77

Kimura
 
T
,
Tse
 
K
,
Sette
 
A
,
Ley
 
K.
 
Vaccination to modulate atherosclerosis
.
Autoimmunity
 
2015
;
48
:
152
160
.

Comments

3 Comments
Infections and Athrothrombotic Disease
27 November 2017
bgs43402@yahoo.com
The Bowling Green Study
I thank Drs Pothineni and Nehta for their response to my comment. I can only comment on my experience in the Western World, but I would point out that we can predict the population at risk of ATD with high accuracy, which means that the organisms cause "pro-inflammatory" infections would have the ability to selectively affect/infect only those with the known ATD risk factors. That being said, I do have a patient who sustained an acute myocardial infarction despite high-intensity statin therapy for moderate dyslipidemia--and he also had systemic sclerosis and at the time of his AMI had pneumonia. In my experience, one should screen for ATD risk factors and treat them to plaque stabilization/regression levels and give aspirin for its anti-platelet effects. This should minimize any inflammatory issues.
Submitted on 27/11/2017 4:16 PM GMT
Inflammation and Atherothrombotic Disease
22 November 2017
Naga Venkata Pothineni, Jawahar L Mehta
University of Arkansas for Medical Sciences
We agree that the inflammatory component of athero thrombosis is multi factorial. Traditional cardiovascular risk factors such as hypertension, diabetes and smoking potentiate the inflammatory milieu that contributes to athero-thrombotic disease. In addition, chronic infections do play a significant role, especially in parts of the world where traditional cardiovascular risk factors are not as prevalent. It may be impossible to include this variable in a risk prediction tool such as the Framingham equation, but does not denounce the clinical importance of pro-atherosclerotic infections
Submitted on 22/11/2017 3:24 PM GMT
Inflammation and Atherothrombotic Disease
17 November 2017
bgs43402@yahoo.com
The Bowling Green Study
The inflammatory component of atherothrombotic disease (ATD) is a reaction to the known ATD risk factors: cigarette smoking, dyslipidemia, hypertension, and diabetes. When Paul Rdiker, MD, enunciated his views on the matter, he pointed out that inflammatory burden is high in subsaharan Africa but ATD risk factors are uncommon, and ATD is uncommon. This work was reproduced in South American Indians. William Kannel, MD, when asked why the Framingham Risk Score did not include family history, responded that in the absence of ATD risk factors, ATD was uncommon.
Submitted on 17/11/2017 6:33 PM GMT