Abstract

As current therapies for cardiovascular disease (CVD), predominantly based on lipid lowering, still face an unacceptable residual risk, novel treatment strategies are being explored. Besides lipids, inflammatory processes play a major role in the pathogenesis of atherosclerosis, the underlying cause of the majority of CVD. The first clinical trials targeting the interleukin-1β-inflammasome axis have shown that targeting this pathway is successful in reducing cardiovascular events but did not decrease overall CVD mortality. Hence, novel and improved immunotherapeutics to treat CVD are being awaited.

Cardiovascular pathologies and immunology: two disciplines born apart?

Immunology and cardiology might seem two different fields of medicine, but they considerably overlay. They stem out of the same fertile soil and only recently researchers started digging into it, clarifying the role of the immune system in cardiovascular pathologies, among which heart failure (HF), myocardial infarction (MI) and atherosclerosis.

Atherosclerosis is defined as a chronic inflammatory disease which determines a slowly progressing formation of luminal plaques in large- and medium-sized arteries. These atherosclerotic plaques may rupture, erode or become unstable leading to fatal adverse events such as ischemic heart disease, stroke and peripheral vascular disease, collectively defined as cardiovascular diseases (CVD). Hence, vascular inflammation participates in atherosclerotic plaque initiation, perpetuation and instability.

Plaque formation is driven by increased blood lipid levels, in particular hypercholesterolemia as well as maladaptive immune responses, determining a state of chronic inflammation in the arterial wall.1 The disease process occurs predominantly at sites of disturbed laminar flow, i.e. arterial branchpoints and bifurcations.2 The turbulent blood flow can cause endothelial injury and a consequent release of inflammatory mediators. In addition, low-density lipoproteins (LDL) start to accumulate in subendothelial layers and get modified (OxLDL). Through these modifications, LDL gets phagocytosed by macrophages, which transform into the so-called ‘foam cells’ forming a luminal fatty streak. (Ox)LDL is antigenic and enhances leukocyte recruitment, including T cells and B cells. Lastly, smooth muscle cells migrate to the surface of the plaque forming a fibrous cap.

The use of lipid-lowering drugs such as statins, the hydroxyl-methyl-glutaryl-CoA (HMG-CoA) reductase inhibitors, and more recently developed proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, has shown promising results in the clinic. Both classes of drugs effectively control LDL cholesterol and reduce major adverse cardiovascular events by almost 50%.3 Nowadays, it has been established that statins also have additional effects. On one hand, they phenotypically determine plaque stabilization by reducing lipid content and a greater fibrous-cap thickness,4 and this change in plaque phenotype has been associated with an increase in the plaque erosion/plaque rupture ratio;5 on the other hand, they have potent anti-inflammatory properties. Moreover, clinical studies, such as the Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) corroborate that inhibition of inflammation through statins affects clinical outcomes, independently of lipid lowering.6

Furthermore, bempedoic acid, a nonstatin antihyperlipidemic drug developed by Esperion, has been approved in 2020 for the treatment of hypercholesterolemia, as monotherapy or in combination with ezetimibe, an inhibitor of intestinal cholesterol absorption.7

However, albeit their effectiveness, a substantial part of the population still suffers from CVD. Hence, during the past decades basic and clinical research have generated a large body of evidence on the possibility of targeting inflammation, the other crucial component contributing to atherosclerosis as a potential therapeutic strategy in the treatment of atherosclerosis. In this review, we highlight the novel avenues for immunotherapeutics in CVD.

Cardiovascular disease and immunotherapy

The journey of immunotherapy in chronic inflammatory diseases commenced by blocking the most prominent inflammatory cytokines involved, being it a cardiovascular, autoimmune or rheumatoid disease. One successful example is the use of targeting tumor necrosis factor (TNF)-α in patients affected by rheumatoid arthritis (RA), a chronic disease that leads to inflammation and progressive joint damage.8 Treatment with anti-TNF-α antibodies has been considered state of the art for many years, albeit some patients did not fully benefit. Interestingly, targeting inflammation not only attenuated inflammation-associated joint damage but also reduced the risk of CV events in RA patients.9

A crucial inflammatory cytokine involved in the pathogenesis is interleukin (IL)-1β, which upregulates downstream inflammatory cytokines, such as TNF-α and IL-6, as well as acute phase reactants, such as C-reactive protein (CRP), fibrinogen and plasminogen activator inhibitor.10 In 2017, the pivotal Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial demonstrated for the first time that targeting the inflammatory IL-1β pathway with the monoclonal antibody canakinumab led to a significant reduction of first recurrent CVD events in patients with prior MI and residual inflammation independent of lipid-lowering therapies.11 Residual inflammation is clinically defined by a persistently elevated (>2 mg/l) high-sensitive C-reactive protein (hs-CRP). hs-CRP concentrations >1 mg/dl (10 mg/l) indicate clinically significant inflammation, whereas concentrations between 0.3 and 1 mg/dl (3 and 10 mg/l; minor CRP elevation) indicate low-grade inflammation, the typical inflammation which characterizes CVD.

In a prespecified secondary analysis, Ridker et al. further provided evidence that IL-1β blockade was capable of reducing CV and all-cause mortality by 31%, but only in patients who achieved on-treatment hs-CRP concentrations below 2 mg/l. In patients with hs-CRP concentrations at 2 mg/l or above, no significant reduction in these endpoints was observed.12

Of note, patients enrolled in the CANTOS trial not only have high risk of a first CV event but also remain at higher risk for recurrent eents. The approach of recording just the first CV event underestimated the disease burden of the patient. Therefore, a more recent subgroup analysis of the CANTOS study done by Everett et al. aimed at covering this issue. Patients randomized for the CANTOS trial incurred to an endpoint were asked to remain in the trial for its total duration. Initial and subsequent CV events were collected, allowing a broader assessment of the efficacy of canakinumab. This new study demonstrated that hampering inflammation through canakinumab in patients with prior MI and evidence of ongoing subclinical inflammation reduces the total number of serious recurrent CV events, including nonfatal MI and stroke, unstable angina requiring unplanned or planned coronary revascularization and CV death.13

Treatment with canakinumab was also effective in lowering levels of IL-6,14 a central pleiotropic inflammatory cytokine involved in the pathogenesis of multiple inflammatory disorders such as RA and psoriasis.15 Overall, results obtained through the CANTOS trial demonstrated that targeting inflammation could substantially prevent atherosclerosis-related adverse CV events.16 However, benefits of treatment have to outweigh the risks. In fact, the attempt of blocking inflammation with canakinumab was accompanied by downsides such as leukopenia, higher incidence of fatal infections or sepsis,11 which mandates for the development of other, safer anti-inflammatory drugs.

Using an alternative anti-inflammatory approach with low-dose methotrexate (15–20 mg weekly), a disease-modifying antirheumatic drugs (DMARDs) previously established to control the clinical activity of RA, did not reduce inflammation in patients with a history of MI or multivessel coronary disease that also had a history of type 2 diabetes or metabolic syndrome. Specifically, among patients with stable atherosclerosis enrolled in the Cardiovascular Inflammation Reduction Trial (CIRT), the treatment with low-dose methotrexate did not reduce levels of IL-1β, IL-6 or CRP and consequently did not result in fewer CV events as compared to placebo.17

Another drug proposed at blocking the underlying inflammatory diathesis in CVD is colchicine, an inexpensive, orally administered, potent anti-inflammatory drug, known to affect the inflammasome via affecting tubulin organization.18 While the first Low Dose Colchicine trial (LoDoCo)19 only involved a smaller patient-cohort (532 patients) and was not placebo-controlled, but open-label trial instead, a second study, the Colchicine Cardiovascular Outcome Trial (COLCOT) was more elaborate, also in assessing long-term outcomes and safety profile of the drug. In both studies, 0.5 mg/day of colchicine, given on top of other secondary-prevention therapeutic strategies, such as aspirin, clopidogrel and statins, was effective in the prevention of ischemic CV events in patients with stable coronary disease and a recent MI, respectively. However, the mechanism(s) by which colchicine exerts its anti-inflammatory CV-related effects still remain obscure. Contrary to canakinumab-treatment, colchicine-treatment did not reduce hs-CRP levels or other plasma markers of inflammation as compared to placebo. In line, the treatment effect on the various components of the combined primary endpoint is rather weak and requires further investigations.20 Nevertheless, results of a further trial named Low Dose Colchicine trial II (LoDoCo II) trial demonstrated that low-dose colchicine improves CV outcomes in patients with clinically stable coronary artery disease (CAD).16 The LoDoCo II trial was, in contrast to the previous mentioned LoDoCo trial an investigator-initiated, randomized, controlled, double-blind, event-driven trial, which however provided confirmation to the LoDoCo trial.21

In synopsis, the strategy of anti-inflammatory targeting has been validated, albeit the partly negative results, still elusive mechanisms and the susceptibility to fatal infections mandate for more specific anti-inflammatory strategies.

It is crucial to bear in mind that inflammation also plays an important role in the pathogenesis of other cardiac pathologies such as heart failure (HF) and the consequent cardiac remodeling.22 HF represents a major cause of hospitalization, morbidity and mortality in developed countries. Similar to the pathogenesis of atherosclerosis, a vicious cycle between inflammation and declining heart function exists. The role of inflammation in HF was firstly recognized by Levine et al.23 in 1990, who reported elevated level of TNF-α in HF patients with reduced ejection fraction (HFrEF) as compared to healthy individuals. Although this inappropriate immune activation and inflammation ought to be a therapeutic target in patients affected by chronic HF, anti-TNF trials with Etanercept, a soluble receptor that binds both TNF-α and TNF-ß, and Infliximab, a monoclonal antibody against TNF-α, yielded disappointing results.24 Contrarily, more encouraging results were obtained in animal models by targeting the adaptive immunity and hence blocking T cell costimulation with the Cytotoxic T lymphocyte antigen 4 (CTLA4) immunoglobulin abatacept. Through inhibition of T cell costimulation, the pathogenesis of HF was delayed and its severity reduced in aging mice.25,26

Inflammation also plays a pivotal role in the pathogenesis of ischemia/reperfusion (I/R) which is a paradoxical exacerbation of cellular damage and death, after the restoration of blood flow to the ischemic tissue. Hypoxia and reperfusion contribute to the expression of several inflammatory cytokines, namely IL-6, TNF-α and IL-1. Animal studies attest the potential beneficial effect of blocking TNF-α for reducing I/R severity. However, a respective clinical trial aimed at blocking TNF-α in human I/R injury has not been reported.27 Interestingly, toll-like receptor 4 (TLR4), which is a sensor of lipopolysaccharide, has been identified to be an additional enhancer of cardiac injury and inflammation in the setting of I/R injury.28 In mice, treatment with the specific TLR4 antagonist eritoran reduced M/R injury as well as markers of inflammation,29 suggesting that TLR4 is an attractive therapeutic target for I/R injury.

In synopsis, immunomodulation after MI seems currently a promising road to unravel. Strategies include blockade of early initiators of inflammation, namely cytokines, chemokines and reactive oxidative species, as well as downstream blockade of the adaptive immune system, including B cells and T cells. Despite the great effort of the scientific community, the plethora of targets, the current timing and the dosage of potential therapies are issues yet to be faced.

The future ahead

Recent clinical trials have proven that inflammation plays an important role in the pathogenesis of human atherosclerosis, and that targeting inflammation for combating CVD has a huge treatment potential (Table 1). Hence, when designing novel effective therapeutic strategies, there is a multitude of options. Here, we will focus on the therapeutic potential of two immunological pathways, that have been proven to be pivotal in atherosclerosis (i) chemokine-induced immune cell recruitment and migration and (ii) immune modulation via costimulatory and coinhibitory immune checkpoints.

Table 1.

Approved anti-inflammatory therapies for cardiovascular disease

Therapeutic agentMechanism of actionEffectsStudy/clinical trial
CanakinumabIL-1β antibodyReduction of inflammation and recurrence of CV eventsCANTOS (Ridker et al.)11
Methothrexate (DMARD)Inhibitor of purine metabolismNo beneficial effectsCIRT (Ridker et al.)17
ColchicineTubulin disruptionPrevention of ischemic CV in patients with stable CAD, no reduction of hs-CRPLoDoCo (Nidorf et al.)19
LoDoCo II (Nidorf et al.)21
COLCOT (Tardif et al.)20
Therapeutic agentMechanism of actionEffectsStudy/clinical trial
CanakinumabIL-1β antibodyReduction of inflammation and recurrence of CV eventsCANTOS (Ridker et al.)11
Methothrexate (DMARD)Inhibitor of purine metabolismNo beneficial effectsCIRT (Ridker et al.)17
ColchicineTubulin disruptionPrevention of ischemic CV in patients with stable CAD, no reduction of hs-CRPLoDoCo (Nidorf et al.)19
LoDoCo II (Nidorf et al.)21
COLCOT (Tardif et al.)20
Table 1.

Approved anti-inflammatory therapies for cardiovascular disease

Therapeutic agentMechanism of actionEffectsStudy/clinical trial
CanakinumabIL-1β antibodyReduction of inflammation and recurrence of CV eventsCANTOS (Ridker et al.)11
Methothrexate (DMARD)Inhibitor of purine metabolismNo beneficial effectsCIRT (Ridker et al.)17
ColchicineTubulin disruptionPrevention of ischemic CV in patients with stable CAD, no reduction of hs-CRPLoDoCo (Nidorf et al.)19
LoDoCo II (Nidorf et al.)21
COLCOT (Tardif et al.)20
Therapeutic agentMechanism of actionEffectsStudy/clinical trial
CanakinumabIL-1β antibodyReduction of inflammation and recurrence of CV eventsCANTOS (Ridker et al.)11
Methothrexate (DMARD)Inhibitor of purine metabolismNo beneficial effectsCIRT (Ridker et al.)17
ColchicineTubulin disruptionPrevention of ischemic CV in patients with stable CAD, no reduction of hs-CRPLoDoCo (Nidorf et al.)19
LoDoCo II (Nidorf et al.)21
COLCOT (Tardif et al.)20

Targeting cell recruitment and migration: chemokines

From ancient Greek, diapedesis is the movement of a cell towards an injured or inflamed site and is a crucial process in atherogenesis. When vascular endothelial cells get activated by turbulent blood flow and oxidative stress, leukocytes start to roll, adhere and migrate into subendothelial tissue. This process is carefully orchestrated by a dyad of chemokines and their receptors, which in turn regulate immune responses.30 As mentioned earlier, during the process of atherosclerotic plaque formation and under hyperlipidemic conditions, LDL becomes oxidized and induces the expression of CXCL1 by endothelial cells. CXCL1 interacts with CXCR2 on myeloid cells, stimulating their recruitment to sites of chronic inflammation. CXCR2 deficient mice and mice with systemic absence of CXCL1 displayed reduced lesion size and macrophage and apoptotic cell content.31 The same holds true when CXCR2 is specifically knocked down in hematopoietic cells.32 Effects in mice deficient for the CXCR2 ligand CXCL1 were shown to be similar, with a greater importance of CXCR2 over CXCL1 for macrophage accumulation in established lesions.33

The CCL2/CCR2 axis is crucial for mediating the mobilization of classical and inflammatory monocytes.34,35 Consequently, in mice depletion of CCR2 resulted in less atherosclerotic lesions.1 Deficiency in both CCL2 and CX3CR1, or CCR2 and CX3CL further reduced plaque burden. Moreover, silencing of CCR2 through siRNA has been shown to be effective in attenuating monocyte mobilization and infarct-related inflammation in mouse models of MI.36 In fact, chemokines involved in leukocyte migration during inflammation, may also contribute to injury or repair of myocardial tissue after MI. Lack of CCR2, receptor for CCL2 in mice, determines decrease in macrophage infiltration and infarct size compared to wild-type mice.37 CXCR2 mediates not only neutrophil chemotaxis but also inhibits cardiomyocyte death induced by hypoxia. Thereupon, its role is controversial on myocardial availability during I/R injury, with a slightly predominant damaging effect over tissue protection.38

Another strategy showed, that the administration of a nonantagonistic chemotactic protein-1 (MCP-1/CCL2) mutant, PA508 reduces inflammatory monocyte recruitment, thereby limiting neointimal hyperplasia, as well as lower ischemia/reperfusion injury in mice.39

In humans, the first clinical trials using chemokine inhibition, have proven the potential for this class of immunotherapeutics for CVD. In a randomized, double-blind, placebo-controlled phase 2a trial (ClinicalTrials.gov Identifier: NCT00715169) using the CCR2 inhibitor MLN1202, a monoclonal antibody which blocks CCR2-CCL2 interactions, reduced CRP levels in patients with atherosclerotic CVD, defined as >2 risk factor for atherosclerotic CVD and circulating CRP levels >3 mg/l).40

CCL5 and CXCL4 chemokine signaling mediates integrin activation and leukocyte arrest on inflamed endothelium. Activated platelets deposit CCL5 on the endothelial surface, with consequent leukocyte recruitment.35 The interaction of CCL5 with its receptor CCR1 and CCR5 requires sialylation hereby creating favorable and improved ligand-receptor interactions. Mice with a deficiency in sialyl-transferase St3Gal-IV display lower monocyte adhesion and atherosclerotic lesion size in a CCL5-related fashion.41 The same outcome is also visible when globally blocking CCR5 in mice using Met-CCL542 or in mice with CCR5 deficiency.43

Accordingly, a Phase 4 clinical trial (ClinicalTrials.gov Identifier: NCT03402815) demonstrated an effective antagonism of CCR5 with the FDA-approved drug maraviroc previously used in HIV infected patients. The administration of the drug (300 mg per os for 24 weeks) combined with antiretroviral therapy (ART) suggested an antiatherosclerotic effect, reflected by improvement of baseline carotid lesions of patients enrolled in the study.44 Those patients were infected with HIV and treated with ART, which both contributed to an increase in atherosclerosis and activation of chronic inflammatory processes45,46 and coinfected with hepatitis C virus (HCV), which may also contribute to increased CVD risk.47

Another pivotal chemokine dyad involved in cell trafficking and immunity is CXCR4-CXCL12, which has a protective role in cells of the arterial wall by sustaining endothelial integrity and promoting contractile properties of vascular smooth muscle cells. In humans, regression analysis identified the C-allele of SNP rs2322864 in the CXCR4 locus to be associated with reduced CXCR4 expression in carotid artery plaques and a higher risk of coronary heart disease. Moreover, reduced expression of CXCR4 in carotid plaques correlated with a higher prevalence of symptomatic disease,48 suggesting that selective modulators of arterial CXCR4 might be a novel therapeutic option to treat atherosclerosis.

In line, a more recent study conducted by Kontos et al. proved the efficacy of a soluble engineered molecule (msR4M-L1) able to selectively block the interaction between macrophage migration inhibitor factor-1 (MIF-1), an atypical chemokine promoting atherosclerosis through CXCR4. This novel strategy showed promising results in hampering the MIF-1/CXCR4 interaction, without disrupting the atheroprotective effect of the CXCR4-CXCL12 axis.49

In conclusion, due to their crucial role in mediating cell trafficking and inflammation, targeting chemokine-chemokine receptor interactions is a promising option to treat atherosclerotic CVD (Table 2A). Albeit the redundancy of the chemokine system, it is of paramount importance to correctly select the target and the drug dosing, which both constitute the main barriers for developing efficacious chemokine-based anti-inflammatory therapies.53

Table 2.

Potential anti-inflammatory therapeutic targets: (A) chemokines, (B) immune checkpoint modulators

AMechanism of actionEffects
Chemokines
CCR2
siRNA (Majmudar et al.)36CCR2 inhibitor, MLN1202 (Clinical trial NCT00715169)(Gilbert et al.)40
Attenuates monocyte mobilization and infarct-related inflammation in mouse with MI Reduced CRP in patients with atherosclerotic CVD
MCP1/CCL2 mutant
Nonantagonistic chemotactic protein PA508 (Liehn et al.)39
Reduction of neointimal hyperplasia
CCR5
CCR5 antagonist Maraviroc (Clinical trial NCT03402815) (Maggi et al.)44
Improvement baseline carotid lesions
MIF-1
soluble engineered molecule (msR4M-L1) by Kontos et al.49
Hampering CXCR4/MIF-1 interaction resulting in atheroprotection
B
Immune checkpointsCTLA4Abatacept(CTLA4-IgG1)(Hsieh et al.)9Significant CVD reduction in anti-TNF-α nonresponders
CD40-CD40LCD40L antibody(Lutgens et al.)50; Schönbeck et al.)51Transformation of unstable plaque into more stable, lipid poor and collagen rich plaques
CD40-TRAF6Small molecule inhibitor(Lutgens et al.)52Stable atherosclerotic plaque phenotype
AMechanism of actionEffects
Chemokines
CCR2
siRNA (Majmudar et al.)36CCR2 inhibitor, MLN1202 (Clinical trial NCT00715169)(Gilbert et al.)40
Attenuates monocyte mobilization and infarct-related inflammation in mouse with MI Reduced CRP in patients with atherosclerotic CVD
MCP1/CCL2 mutant
Nonantagonistic chemotactic protein PA508 (Liehn et al.)39
Reduction of neointimal hyperplasia
CCR5
CCR5 antagonist Maraviroc (Clinical trial NCT03402815) (Maggi et al.)44
Improvement baseline carotid lesions
MIF-1
soluble engineered molecule (msR4M-L1) by Kontos et al.49
Hampering CXCR4/MIF-1 interaction resulting in atheroprotection
B
Immune checkpointsCTLA4Abatacept(CTLA4-IgG1)(Hsieh et al.)9Significant CVD reduction in anti-TNF-α nonresponders
CD40-CD40LCD40L antibody(Lutgens et al.)50; Schönbeck et al.)51Transformation of unstable plaque into more stable, lipid poor and collagen rich plaques
CD40-TRAF6Small molecule inhibitor(Lutgens et al.)52Stable atherosclerotic plaque phenotype
Table 2.

Potential anti-inflammatory therapeutic targets: (A) chemokines, (B) immune checkpoint modulators

AMechanism of actionEffects
Chemokines
CCR2
siRNA (Majmudar et al.)36CCR2 inhibitor, MLN1202 (Clinical trial NCT00715169)(Gilbert et al.)40
Attenuates monocyte mobilization and infarct-related inflammation in mouse with MI Reduced CRP in patients with atherosclerotic CVD
MCP1/CCL2 mutant
Nonantagonistic chemotactic protein PA508 (Liehn et al.)39
Reduction of neointimal hyperplasia
CCR5
CCR5 antagonist Maraviroc (Clinical trial NCT03402815) (Maggi et al.)44
Improvement baseline carotid lesions
MIF-1
soluble engineered molecule (msR4M-L1) by Kontos et al.49
Hampering CXCR4/MIF-1 interaction resulting in atheroprotection
B
Immune checkpointsCTLA4Abatacept(CTLA4-IgG1)(Hsieh et al.)9Significant CVD reduction in anti-TNF-α nonresponders
CD40-CD40LCD40L antibody(Lutgens et al.)50; Schönbeck et al.)51Transformation of unstable plaque into more stable, lipid poor and collagen rich plaques
CD40-TRAF6Small molecule inhibitor(Lutgens et al.)52Stable atherosclerotic plaque phenotype
AMechanism of actionEffects
Chemokines
CCR2
siRNA (Majmudar et al.)36CCR2 inhibitor, MLN1202 (Clinical trial NCT00715169)(Gilbert et al.)40
Attenuates monocyte mobilization and infarct-related inflammation in mouse with MI Reduced CRP in patients with atherosclerotic CVD
MCP1/CCL2 mutant
Nonantagonistic chemotactic protein PA508 (Liehn et al.)39
Reduction of neointimal hyperplasia
CCR5
CCR5 antagonist Maraviroc (Clinical trial NCT03402815) (Maggi et al.)44
Improvement baseline carotid lesions
MIF-1
soluble engineered molecule (msR4M-L1) by Kontos et al.49
Hampering CXCR4/MIF-1 interaction resulting in atheroprotection
B
Immune checkpointsCTLA4Abatacept(CTLA4-IgG1)(Hsieh et al.)9Significant CVD reduction in anti-TNF-α nonresponders
CD40-CD40LCD40L antibody(Lutgens et al.)50; Schönbeck et al.)51Transformation of unstable plaque into more stable, lipid poor and collagen rich plaques
CD40-TRAF6Small molecule inhibitor(Lutgens et al.)52Stable atherosclerotic plaque phenotype

Targeting adaptive immunity: immune checkpoints

Immunotherapy targeting the adaptive immune system has recently revolutionized the world of cancer therapy, considerably improving patients’ survival, with many drugs targeting immune checkpoints in preclinical and clinical trials, aimed at improving immune responses against cancer cells to promote their killing.54 These results reflect the strong therapeutic potential of this class of immune modulators. Although targeting immune checkpoints has been rapidly integrated in oncology treatment, its exploitation in CVD is still in a preclinical stage.55 The two largest classes of immune checkpoints are costimulatory and coinhibitory molecules, and are both master regulators of immune responses, classically known as ‘signal 2’ that promotes or dampens T-cell activation and proliferation upon T-cell antigen presenting cell interactions,56 but nowadays known to play a pivotal role in the communication and activation of a plethora of cell types, including immune and nonimmune cells.57

Many costimulatory and coinhibitory molecules, including the CD80/86-CD28/cytotoxic T lymphocyte-associated antigen 4 (CTLA4), CD40-CD40L, CD27-CD70, OX40L-OX40, GITR-GITRL and PD1-PDL1/2 axis have been shown to mediate atherogenesis in a laboratory setting57 (Table 2B). Only recently, the first clinical studies, conducted in other diseases, have revealed the potential of targeting these immune checkpoints in human CVD.

Abatacept, an IgG1-CTLA4 fusion protein that blocks CD80/86 mediated costimulation is an FDA approved drug indicated for the treatment of autoimmune diseases and particularly effective in RA patients that respond poorly to methotrexate or anti-TNF-α treatment.58

Data obtained from RA trials show a beneficial effect of abatacept on CVD outcomes. In a study comparing biologics with synthetic DMARDs, abatacept treatment resulted in a significant reduction of CVD (HR 0.5, 95% CI 0.30–0.83).59 Also, abatacept reduced CVD risk in anti-TNF-α nonresponders in a 2-year follow-up in MI, MACE, stroke and HF compared to rituximab users.9 Similar results were found in a study where RA patients using TNF-α inhibitors were compared to abatacept users and abatacept was associated with a 20% reduced risk of MACE.60 However, clinical trials testing the effect abatacept on CVD outcomes directly are still being awaited.

Antibodies that antagonize the coinhibitory CTLA4 and/or programmed cell death protein 1 (Ligand) (PD1(L)), the so-called immune checkpoint inhibitors, are a first-in-line treatment for an increasing number of malignancies. Immune checkpoint inhibitors release the brake on T cell activation, elicit strong antitumor immune responses, but also cause immune-related adverse events.61,62 Cardiovascular complications are not uncommon after ICI therapy63,64 and a recent study among 2842 patients who received ICI treatment or not reported that the incidence of atherosclerotic CV events, defined as a composite of MI, coronary revascularization and ischemic stroke, was increased 4.7-fold after ICI therapy.65 These studies not only show the potent role of CTLA4 and PD(L)1 as protectors of CVD but also increase awareness of the detrimental side effects of this highly potent anticancer immunotherapy.

Another costimulatory immune checkpoint that can be considered a potent therapeutic target in CVD is the CD40L-CD40 dyad. In an experimental setting, inhibition of CD40L or CD40, even when inhibited in established atherosclerosis, is highly effective in reducing atherosclerosis, and generates plaques that are rich in collagen and contain a limited number of immune cells, the murine equivalent of a stable, clinically safe, plaque.50–52,66 Although blocking CD40L or CD40 in human CVD seems evident as a potent immunotherapy for CVD, this has never been evaluated in clinical trials. However, antagonistic CD40L and CD40 antibodies, such as CDP7657, VIB4920 and iscalimab are available and have been successfully tested in a plethora of other chronic inflammatory diseases, such as multiple sclerosis, Sjogren syndrome, SLE and transplant rejection, and the outcomes of phase II and III trials are being awaited and therefore, effects on CVD outcomes in these study populations are not yet available. However, preclinical studies, with CD40L and CD40 antagonists designed to target atherosclerotic CVD, have been performed. CD40 does not have intrinsic signal capabilities and needs adaptor molecules, the TNF-receptor associated factors (TRAFs), to exert signaling. Using mice specifically lacking CD40-TRAF6 or CD40-TRAF2/3 interactions, it was shown that only mice deficient in CD40-TRAF6 were protected against atherosclerosis,52 show that especially CD40-TRAF6 interactions, that predominantly take place in macrophages, drive atherosclerosis. A small-molecule inhibitor (SMI) that targets CD40-TRAF6 signaling52,67 was designed, and was shown to reduce (existing) atherosclerosis in mice, and induces a stable atherosclerotic plaque phenotype, without causing immune-suppressive or thrombo-embolic side effects.68 This SMI’s specific delivery to macrophages using HDL nanobiologics stabilized atherosclerotic plaques in mice and was proven safe in nonhuman primates69 and is currently being developed for in-human treatment. A plethora of preclinical data shows that antagonizing the CD40L-CD40 pathway has a true potential as an immunotherapeutic target to treat CVD. Phases I–III trials have revealed that targeting CD40L or CD40 in humans is safe and well-tolerized.

Synopsis

The increasing interest in the field of cardio-immunology is fueled by the growing awareness of the role of inflammation in cardiovascular pathologies, especially atherosclerosis. Although the CANTOS trial has provided evidence of lowered MI-, stroke- and cardiovascular risk, treatment with canakinumab did not reduce overall CV mortality. Given the adverse events after treatment with canakinumab, immune functions deleterious in the cardiovascular system may be necessary for host defense. Hence, novel immunotherapeutic targets need to be developed and tested for their potential and safety in CVD. Based on a large amount of preclinical and scattered clinical data, targeting chemokines or immune checkpoints might result in a better and more safe reduction of atherosclerosis-associated CVD. Although the time is right to introduce immunotherapy as a valid treatment option for CVD, one always needs to bear in mind that immunotherapy itself in CVD is a double-edged sword, and that therapeutics should be tailored in terms of targets and drug delivery.

Funding

This study was supported by the Deutsche Forschungsgemeinschaft [Grant numbers DFG Einzelförderung 451372580 to D.A.; CRC 1123 to D.A., E.L., C.W.; INST409/150-1FUGG to C.W.] and the German Center for Cardiovascular Research (DZHK) [Promotion of Women Scientist grant 81X1600210 to D.A.]. We also acknowledge the support from the Netherlands CardioVascular Research Initiative: the Dutch Heart Foundation, Dutch Federation of University Medical Centres, the Netherlands Organization for Health Research and Development and the Royal Netherlands Academy of Sciences for the GENIUS-II project “Generating the best evidence-based pharmaceutical targets for atherosclerosis” [CVON2017-20]. This study was also supported by the Netherlands Organization for Scientific Research (NWO) [VICI grant to E.L.]; the EU [Horizon 2020, REPROGRAM to E.L.] and the European Research Council [ERC consolidator grant to E.L, ERC advanced grant to C.W.]. C.W. is a Van de Laar professor of atherosclerosis.

Conflict of interest. The authors have no relevant financial or non-financial interests to disclose.

References

1

Weber
C
,
Noels
H.
Atherosclerosis: current pathogenesis and therapeutic options
.
Nat Med
2011
;
17
:
1410
22
.

2

Moore
KJ
,
Tabas
I.
Macrophages in the pathogenesis of atherosclerosis
.
Cell
2011
;
145
:
341
55
.

3

Ridker
PM
,
Danielson
E
,
Fonseca
FA
,
Genest
J
,
Gotto
AM
,
Kastelein
JJP
, et al.
Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein
.
N Engl J Med
2009
;
64
:
168
70
.

4

Takano
M
,
Mizuno
K
,
Yokoyama
S
,
Seimiya
K
,
Ishibashi
F
,
Okamatsu
K
, et al.
Changes in coronary plaque color and morphology by lipid-lowering therapy with atorvastatin: serial evaluation by coronary angioscopy
.
J Am Coll Cardiol
2003
;
42
:
680
6
.

5

Fahed
AC
,
Jang
IK.
Plaque erosion and acute coronary syndromes: phenotype, molecular characteristics and future directions
.
Nat Rev Cardiol
2021
;
0123456789
.

6

Ridker
PM
,
Danielson
E
,
Fonseca
FA
,
Genest
J
,
Gotto
AM
,
Kastelein
JJ
, et al.
Reduction in C-reactive protein and LDL cholesterol and cardiovascular event rates after initiation of rosuvastatin: a prospective study of the JUPITER trial
.
Lancet
2009
;
373
:
1175
82
.

7

Markham
A.
Bempedoic acid: first approval
.
Drugs
2020
;
80
:
747
53
.

8

Choy EH, Panayi GS
.
Cytokine pathways and joint inflammation in rheumatoid arthritis
.
N Engl J Med
2001
;
344
:
907
16
.

9

Atzeni
F
,
Rodríguez-Carrio
J
,
Popa
CD
,
Nurmohamed
MT
,
Szűcs
G
,
Szekanecz
Z.
Biologic agents reduce cardiovascular events in rheumatoid arthritis not responsive to tumour necrosis factor inhibitors: a national cohort study
.
Can J Cardiol
2021
;
17
:
270
46
.

10

Libby
P.
Interleukin-1 beta as a target for atherosclerosis therapy: biological basis of CANTOS and beyond
.
J Am Coll Cardiol
2017
;
70
:
2278
89
.

11

Ridker
PM
,
Everett
BM
,
Thuren
T
,
MacFadyen
JG
,
Chang
WH
,
Ballantyne
C
, et al. ;
CANTOS Trial Group
.
Antiinflammatory therapy with canakinumab for atherosclerotic disease
.
N Engl J Med
2017
;
377
:
1119
31
.

12

Ridker
PM
,
MacFadyen
JG
,
Everett
BM
,
Libby
P
,
Thuren
T
,
Glynn
RJ
, et al.
Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial
.
Lancet
2018
;
391
:
319
28
.

13

Everett
BM
,
MacFadyen
JG
,
Thuren
T
,
Libby
P
,
Glynn
RJ
,
Ridker
PM.
Inhibition of interleukin-1β and reduction in atherothrombotic cardiovascular events in the CANTOS trial
.
J Am Coll Cardiol
2020
;
76
:
1660
70
.

14

Ridker
PM
,
Libby
P
,
MacFadyen
JG
,
Thuren
T
,
Ballantyne
C
,
Fonseca
F
, et al.
Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: analyses from the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS)
.
Eur Heart J
2018
;
39
:
3499
507
.

15

Scheller
J
,
Chalaris
A
,
Schmidt-Arras
D
,
Rose-John
S.
The pro- and anti-inflammatory properties of the cytokine interleukin-6
.
Biochim Biophys Acta - Mol Cell Res
2011
;
1813
:
878
88
.

16

Weber
C
,
von Hundelshausen
P.
CANTOS trial validates the inflammatory pathogenesis of atherosclerosis
.
Circ Res
2017
;
121
:
1119
21
.

17

Ridker
PM
,
Everett
BM
,
Pradhan
A
,
MacFadyen
JG
,
Solomon
DH
,
Zaharris
E
, et al. ;
CIRT Investigators
.
Low-dose methotrexate for the prevention of atherosclerotic events
.
N Engl J Med
2019
;
380
:
752
62
.

18

Ben-Chetrit
E
,
Levy
M.
Colchicine: 1998 update
.
Semin Arthritis Rheum
1998
;
28
:
48
59
.

19

Nidorf
SM
,
Eikelboom
JW
,
Budgeon
CA
,
Thompson
PL.
Low-dose colchicine for secondary prevention of cardiovascular disease
.
J Am Coll Cardiol
2013
;
61
:
404
10
.

20

Tardif
J-C
,
Kouz
S
,
Waters
DD
,
Bertrand
OF
,
Diaz
R
,
Maggioni
AP
, et al.
Efficacy and safety of low-dose colchicine after myocardial infarction
.
N Engl J Med
2019
;
381
:
2497
505
.

21

Nidorf
SM
,
Fiolet
ATL
,
Mosterd
A
,
Eikelboom
JW
,
Schut
A
,
Opstal
TSJ
, et al.
Colchicine in patients with chronic coronary disease
.
N Engl J Med
2020
;
383
:
1838
47
.

22

Prabhu
SD
,
Frangogiannis
NG.
The biological basis for cardiac repair after myocardial infarction
.
Circ Res
2016
;
119
:
91
112
.

23

Levine
B
,
Kalman
J
,
Mayer
L
,
Fillit
HM
,
Packer
M.
Elevated circulating levels of tumor necrosis factor in severe chronic heart failure
.
New Engl J Med
1990
;
323
:
1120
3
.

24

Mann
DL
,
McMurray
JJV
,
Packer
M
,
Swedberg
K
,
Borer
JS
,
Colucci
WS
, et al.
Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL)
.
Circulation
2004
;
109
:
1594
602
.

25

Martini
E
,
Cremonesi
M
,
Panico
C
,
Carullo
P
,
Bonfiglio
CA
,
Serio
S
, et al.
T cell costimulation blockade blunts age-related heart failure
.
Circ Res
2020
;
127
:
1115
7
.

26

Kallikourdis
M
,
Martini
E
,
Carullo
P
,
Sardi
C
,
Roselli
G
,
Greco
CM
, et al.
T cell costimulation blockade blunts pressure overload-induced heart failure
.
Nat Commun
2017
;
8
:
14680
7
.

27

Fitridge
R
,
Cowled
P.
In:
Fitridge
R
,
Thompson
M
, eds.
Mechanism of Vascular Disease: A Reference Book for Vascular Specialists, Chapter 18
[Internet].
Barr Smith Press
, Adelaide (AU): University of Adelaide Press;
2011
, List of Contributors
331
45
.

28

Lee
SM
,
Hutchinson
M
,
Saint
DA.
The role of toll-like receptor 4 (TLR4) in cardiac ischaemic-reperfusion injury, cardioprotection and preconditioning
.
Clin Exp Pharmacol Physiol
2016
;
43
:
864
71
.

29

Shimamoto
A
,
Chong
AJ
,
Yada
M
,
Shomura
S
,
Takayama
H
,
Fleisig
AJ
, et al.
Inhibition of toll-like receptor 4 with eritoran attenuates myocardial ischemia-reperfusion injury
.
Circulation
2006
;
114
:
270
4
.

30

Zernecke
A
,
Weber
C.
Chemokines in the vascular inflammatory response of atherosclerosis
.
Cardiovasc Res
2010
;
86
:
192
201
.

31

Soehnlein
O
,
Drechsler
M
,
Döring
Y
,
Lievens
D
,
Hartwig
H
,
Kemmerich
K
, et al.
Distinct functions of chemokine receptor axes in the atherogenic mobilization and recruitment of classical monocytes
.
EMBO Mol Med
2013
;
5
:
471
81
.

32

Boisvert
WA
,
Santiago
R
,
Curtiss
LK
,
Terkeltaub
RA.
A leukocyte homologue of the IL-8 receptor CXCR-2 mediates the accumulation of macrophages in atherosclerotic lesions of LDL receptor- deficient mice
.
J Clin Invest
1998
;
101
:
353
63
.

33

Boisvert
WA
,
Rose
DM
,
Johnson
KA
,
Fuentes
ME
,
Lira
SA
,
Curtiss
LK
, et al.
Up-regulated expression of the CXCR2 ligand KC/GRO-α in atherosclerotic lesions plays a central role in macrophage accumulation and lesion progression
.
Am J Pathol
2006
;
168
:
1385
95
.

34

Serbina
NV
,
Pamer
EG.
Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2
.
Nat Immunol
2006
;
7
:
311
7
.

35

Drechsler
M
,
Duchene
J
,
Soehnlein
O.
Chemokines control mobilization, recruitment, and fate of monocytes in atherosclerosis
.
Arterioscler Thromb Vasc Biol
2015
;
35
:
1050
5
.

36

Majmudar
MD
,
Keliher
EJ
,
Heidt
T
,
Leuschner
F
,
Truelove
J
,
Sena
BF
, et al.
Monocyte-directed RNAi targeting CCR2 improves infarct healing in atherosclerosis-prone mice
.
Circulation
2013
;
127
:
2038
46
.

37

Kaikita
K
,
Hayasaki
T
,
Okuma
T
,
Kuziel
WA
,
Ogawa
H
,
Takeya
M.
Targeted deletion of CC chemokine receptor 2 attenuates left ventricular remodeling after experimental myocardial infarction
.
Am J Pathol
2004
;
165
:
439
47
.

38

Tarzami
ST
,
Miao
W
,
Mani
K
,
Lopez
L
,
Factor
SM
,
Berman
JW
, et al.
Opposing effects mediated by the chemokine receptor CXCR2 on myocardial ischemia-reperfusion injury: recruitment of potentially damaging neutrophils and direct myocardial protection
.
Circulation
2003
;
108
:
2387
92
.

39

Liehn
EA
,
Piccinini
AM
,
Koenen
RR
,
Soehnlein
O
,
Adage
T
,
Fatu
R
, et al.
A new monocyte chemotactic protein-1/chemokine cc motif ligand-2 competitor limiting neointima formation and myocardial ischemia/reperfusion injury in mice
.
J Am Coll Cardiol
2010
;
56
:
1847
57
.

40

Gilbert
J
,
Lekstrom-Himes
J
,
Donaldson
D
,
Lee
Y
,
Hu
M
,
Xu
J
, et al. ;
MLN1202 Study Group
.
Effect of CC chemokine receptor 2 CCR2 blockade on serum C-reactive protein in individuals at atherosclerotic risk and with a single nucleotide polymorphism of the monocyte chemoattractant protein-1 promoter region
.
Am J Cardiol
2011
;
107
:
906
11
.

41

Döring
Y
,
Noels
H
,
Mandl
M
,
Kramp
B
,
Neideck
C
,
Lievens
D
, et al.
Deficiency of the sialyltransferase St3Gal4 reduces Ccl5-mediated myeloid cell recruitment and arrest
.
Circ Res
2014
;
114
:
976
81
.

42

Veillard
NR
,
Kwak
B
,
Pelli
G
,
Mulhaupt
F
,
James
RW
,
Proudfoot
AEI
, et al.
Antagonism of RANTES receptors reduces atherosclerotic plaque formation in mice
.
Circ Res
2004
;
94
:
253
61
.

43

Braunersreuther
V
,
Zernecke
A
,
Arnaud
C
,
Liehn
EA
,
Steffens
S
,
Shagdarsuren
E
, et al.
Ccr5 but not Ccr1 deficiency reduces development of diet-induced atherosclerosis in mice
.
Arterioscler Thromb Vasc Biol
2007
;
27
:
373
9
.

44

Maggi
P
,
Bruno
G
,
Perilli
F
,
Saracino
A
,
Volpe
A
,
Santoro
C
, et al.
Effects of therapy with maraviroc on the carotid intima media thickness in HIV-1/HCV co-infected patients
.
In Vivo (Brooklyn)
2017
;
31
:
125
32
.

45

Freiberg
MS
,
Chang
CCH
,
Kuller
LH
,
Skanderson
M
,
Lowy
E
,
Kraemer
KL
, et al.
HIV infection and the risk of acute myocardial infarction
.
JAMA Intern Med
2013
;
173
:
614
22
.

46

Durand
M
,
Sheehy
O
,
Baril
JG
,
Lelorier
J
,
Tremblay
CL.
Association between HIV infection, antiretroviral therapy, and risk of acute myocardial infarction: a cohort and nested case-control study using Québec’s Public Health Insurance database
.
J Acquir Immune Defic Syndr
2011
;
57
:
245
53
.

47

Fernández-Montero
JV
,
Barreiro
P
,
De Mendoza
C
,
Labarga
P
,
Soriano
V.
Hepatitis C virus coinfection independently increases the risk of cardiovascular disease in HIV-positive patients
.
J Viral Hepat
2016
;
23
:
47
52
.

48

Döring
Y
,
Noels
H
,
Van Der Vorst
EPC
,
Neideck
C
,
Egea
V
,
Drechsler
M
, et al.
Vascular CXCR4 limits atherosclerosis by maintaining arterial integrity: evidence from mouse and human studies
.
Circulation
2017
;
136
:
388
403
.

49

Kontos
C
,
El Bounkari
O
,
Krammer
C
,
Sinitski
D
,
Hille
K
,
Zan
C
, et al.
Designed CXCR4 mimic acts as a soluble chemokine receptor that blocks atherogenic inflammation by agonist-specific targeting
.
Nat Commun
2020
;
11
:
5981
18
.

50

Lutgens
E
,
Cleutjens
KBJM
,
Heeneman
S
,
Koteliansky
VE
,
Burkly
LC
,
Daemen
MJAP.
Both early and delayed anti-CD40L antibody treatment induces a stable plaque phenotype
.
Proc Natl Acad Sci USA
2000
;
97
:
7464
9
.

51

Schönbeck
U
,
Sukhova
GK
,
Shimizu
K
,
Mach
F
,
Libby
P.
Inhibition of CD40 signaling limits evolution of established atherosclerosis in mice
.
Proc Natl Acad Sci USA
2000
;
97
:
7458
63
.

52

Lutgens
E
,
Lievens
D
,
Beckers
L
,
Wijnands
E
,
Soehnlein
O
,
Zernecke
A
, et al.
Deficient CD40-TRAF6 signaling in leukocytes prevents atherosclerosis by skewing the immune response toward an antiinflammatory profile
.
J Exp Med
2010
;
207
:
391
404
.

53

Schall
TJ
,
Proudfoot
AEI.
Overcoming hurdles in developing successful drugs targeting chemokine receptors
.
Nat Rev Immunol
2011
;
11
:
355
63
.

54

Peggs
KS
,
Quezada
SA
,
Allison
JP.
Cancer immunotherapy: co-stimulatory agonists and co-inhibitory antagonists
.
Clin Exp Immunol
2009
;
157
:
9
19
.

55

Lutgens
E
,
Atzler
D
,
Döring
Y
,
Duchene
J
,
Steffens
S
,
Weber
C.
Immunotherapy for cardiovascular disease
.
Eur Heart J
2019
;
40
:
3937
46
.

56

Croft
M.
Co-stimulatory members of the TNFR family: keys to effective T-cell immunity?
Nat Rev Immunol
2003
;
3
:
609
20
.

57

Kusters
PJH
,
Lutgens
E
,
Seijkens
TTP.
Exploring immune checkpoints as potential therapeutic targets in atherosclerosis
.
Cardiovasc Res
2018
;
114
:
368
77
.

58

Genovese
MC
,
Becker
J-C
,
Schiff
M
,
Luggen
M
,
Sherrer
Y
,
Kremer
J
, et al.
Abatacept for rheumatoid arthritis refractory to tumor necrosis factor α Inhibition
.
N Engl J Med
2005
;
353
:
1114
23
.

59

Ozen
G
,
Pedro
S
,
Michaud
K.
The risk of Cardiovascular events associated with disease-modifying anti-rheumatic drugs in rheumatoid arthritis
.
J Rheumatol
2021
;
48
:
648
55
.

60

Kang
EH
,
Jin
Y
,
Brill
G
,
Lewey
J
,
Patorno
E
,
Desai
RJ
, et al.
Comparative cardiovascular risk of abatacept and tumor necrosis factor inhibitors in patients with rheumatoid arthritis with and without diabetes mellitus: a multidatabase cohort study
.
J Am Heart Assoc
2018
;
7
:
1
15
.

61

Baxi
S
,
Yang
A
,
Gennarelli
RL
,
Khan
N
,
Wang
Z
,
Boyce
L
, et al.
Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis
.
BMJ
2018
;
360
:
k793
.

62

Patrinely
JR
,
Young
AC
,
Quach
H
,
Williams
GR
,
Ye
F
,
Fan
R
, et al.
Survivorship in immune therapy: assessing toxicities, body composition and health-related quality of life among long-term survivors treated with antibodies to programmed death-1 receptor and its ligand
.
Eur J Cancer
2020
;
135
:
211
20
.

63

Totzeck
M
,
Lutgens
E
,
Neilan
TG.
Are we underestimating the potential for cardiotoxicity related to immune checkpoint inhibitors?
Eur Heart J
2021
;
42
:
1632
4
.

64

De Silva
P
,
Aiello
M
,
Gu-Trantien
C
,
Migliori
E
,
Willard-Gallo
K
,
Solinas
C.
Targeting CTLA-4 in cancer: is it the ideal companion for PD-1 blockade immunotherapy combinations?
Int J Cancer
2020
;
149
:
31
11
.

65

Drobni
ZD
,
Alvi
RM
,
Taron
J
,
Zafar
A
,
Murphy
SP
,
Rambarat
PK
, et al.
Association between immune checkpoint inhibitors with cardiovascular events and atherosclerotic plaque
.
Circulation
2020
;
142
:
2299
311
.

66

Mach
F
,
Schönbeck
U
,
Sukhova
GK
,
Atkinson
E
,
Libby
P.
Reduction of atherosclerosis in mice by inhibition of CD40 signalling
.
Nature
1998
;
394
:
200
3
.

67

Chatzigeorgiou
A
,
Seijkensd
T
,
Zarzyckae
B
,
Engelf
D
,
Poggif
M
,
S van den
B
, et al.
Blocking CD40-TRAF6 signalling is a therapeutic target in obesity associated insulin resistance
.
Proc Natl Acad Sci USA
2014
;
111
:
4644
.

68

Seijkens
TTP
,
van Tiel
CM
,
Kusters
PJH
,
Atzler
D
,
Soehnlein
O
,
Zarzycka
B
, et al.
Targeting CD40-induced TRAF6 signaling in macrophages reduces atherosclerosis
.
J Am Coll Cardiol
2018
;
71
:
527
42
.

69

Lameijer
M
,
Binderup
T
,
Van Leent
MMT
,
Senders
ML
,
Fay
F
,
Malkus
J
, et al.
Efficacy and safety assessment of a TRAF6-targeted nanoimmunotherapy in atherosclerotic mice and non-human primates
.
Nat Biomed Eng
2018
;
2
:
279
92
.

Author notes

D. Atzler and E. Lutgens contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/pages/standard-publication-reuse-rights)