Skip to main content
Therapeutic Advances in Vaccines logoLink to Therapeutic Advances in Vaccines
. 2017 Feb 1;5(2):39–47. doi: 10.1177/2051013617693753

Vaccine against arteriosclerosis: an update

Kuang-Yuh Chyu 1, Paul C Dimayuga 2, Prediman K Shah 3,
PMCID: PMC5418889  PMID: 28515939

Abstract

Substantial data from experimental and clinical investigation support the role of immune-mediated mechanisms in atherogenesis, with immune systems responding to many endogenous and exogenous antigens that play either proatherogenic or atheroprotective roles. An active immunization strategy against many of these antigens could potentially alter the natural history of atherosclerosis. This review mainly focuses on the important studies on the search for antigens that have been tested in vaccine formulations to reduce atherosclerosis in preclinical models. It will also address the opportunities and challenges associated with potential clinical application of this novel therapeutic paradigm.

Keywords: apolipoprotein B-100, atherosclerosis, immunization, low-density lipoprotein

Background

Atherosclerosis is currently viewed as an immune-mediated inflammatory disease of the arterial wall, with both the innate and adaptive immune systems responding to many endogenous and exogenous antigens. Cells of both innate and adaptive immune systems such as macrophages, dendritic cells, B and T-lymphocytes, and mast cells are involved in atherogenesis, as are the immune-inflammatory mediators such as pathogen-associated and danger-associated molecular pattern molecules, immunoglobulins, cytokines, chemokines, and complement proteins. It is beyond the scope of this review to describe the complex roles of these immune cells and mediators in detail and many excellent reviews are available for the interested readers.15

Given that atherosclerosis is an immune-mediated disease of the arterial wall, it is tempting to consider specific strategies to modulate the immune responses to favorably affect the natural history of atherosclerosis. The challenge of this approach is to identify specific antigens relevant to atherogenesis so that they can be used to activate an antigen-specific atheroprotective immune response. Most studies in search of potential antigens are in preclinical experimental stages; hence data discussed in this review refer to data from animal models of atherosclerosis unless otherwise stated.

From low-density lipoprotein to apoB-100-derived peptides as antigens: our experience

Because low-density lipoprotein (LDL) and other apoB-100-containing lipoproteins are the primary culprits with the strongest causative link with atherosclerosis, investigators have been asking if immunizing hypercholesterolemic animals with LDL or apoB-100-derived peptide-antigens would modulate atherosclerosis. When homologous whole native or modified LDL was used as an antigen in a vaccine formulation, immunizing experimental animals with these vaccine formulations demonstrated atheroprotective effects.614

Because LDL is a large, heterogeneous molecule containing apolipoproteins, cholesteryl esters, triglycerides, and phospholipids, it would be impractical to use whole homologous LDL as an antigen in a clinically usable vaccine formulation. To identify the potential atheroprotective antigenic epitopes in LDL, our laboratory in collaboration with Dr Jan Nilsson’s laboratory at Lund University in Sweden, generated a library of 302 peptides spanning the entire 4536-amino acid sequence in human apoB-100 protein and selected 102 peptides based on the humoral immune response detected in pooled human plasma as potential candidates for the next round of screening.15 Among these 102 peptides, certain peptide sequences, labeled here as p2, p143, and p210 resulted in a 40–70% decrease in atherosclerosis and reduction in plaque inflammation when used in a vaccine formulation in hypercholesterolemic mice.16,17 Knowing the defined epitope makes more definitive immunologic studies possible; our teams have been using p210 as a prototype antigen in vaccine formulations due to its consistent atheroprotective effects.1820

Mechanisms of action of p210 vaccine

Immunization with the p210 vaccine resulted in a significant reduction of aortic atherosclerosis compared with controls in murine model of atherosclerosis.18 Given that immunization activates both B and T-cells,18 it is important to delineate the subset(s) of lymphocytes which mediate the p210 vaccine’s atheroprotective effect. Currently existing experimental evidence does not support a strong role for the humoral response in mediating the protective effect of active immunization using the p210 vaccine. This is based on the observations that (1) apoB-100 peptide immunization reduced atherosclerosis without an increase in peptide-specific immunoglobulin (Ig)G,19 and the induced antibody titers did not correlate with the lesion size;20 (2) Adoptive transfer of B-cells from p210-immunized mice to nonimmunized recipient mice did not confer atheroprotective effect.18 However, immunization with p210 peptide did alter the natural history of p210 antibody levels in apoE-/- mice. In control mice, p210 IgG titer remained low between baseline and 25 weeks; whereas mice receiving adjuvant only or p210 vaccine developed high p210 IgG titer at 25 weeks. Interestingly IgG titer at 25 weeks from p210-immunized mice was lower when compared with that of mice receiving adjuvant only.18 p210 IgM has a different profile; low level before immunization but titers increased over time, regardless of whether or not mice were immunized with p210 vaccine. This observation suggests that: (1) an endogenous IgM immune response against p210 exists, (2) induction of p210 IgG may serve as a marker of vaccination effect but not a marker of atheroprotective efficacy of the p210 vaccine.

Given that immunization with p210 vaccine activated CD8+ T-cells,18 we tested whether CD8+ T-cells could be the immune cells that mediate the vaccine’s atheroprotective effect by adoptively transferring CD8+ T-cells from p210 immunized mice into nonimmunized mice. Such transfer recapitulated the atheroprotective effect of active immunization, confirming that CD8+ T-cells mediate the atheroprotective effect of p210 vaccine.18 Additionally we demonstrated reduction of CD11c+ cells in the plaques, injection site, and draining lymph nodes of p210 immunized mice and effector CD8+ T-cells from p210-immunized mice developed a preferentially higher cytolytic response against p210-loaded dendritic cells in vitro indicating antigen-specific modulation of dendritic cells by p210 vaccine.18 This may explain the observed reduction in dendritic cells in the immunization sites and in atherosclerotic plaques.

Immunization with p210 peptide also elicited a CD4+ T-cell response. The reduction of atherosclerosis by p210 immunization was associated with a CD4+CD25+ T-cell response. Administration of antibodies against CD25 reduced CD4+CD25+ T-cells and abrogated the atheroprotective effect of p210 immunization.21 Immunization of female apoE-/- mice intranasally with a recombinant protein consisting of p210 fused with the cholera toxin B (CTB) subunit (p210-CTB) reduced atherosclerosis in aortic sinuses of mice when compared with control mice. The rationale of using CTB conjugated with p210 was based on (1) CTB promotes the uptake of the antigen via the nasal mucosa to elicit protective immunity; (2) CTB-based vaccines have now been tested in a first human phase II trial in Behcet’s disease. The investigators also observed that splenic CD4+ T-cells from p210-CTB-immunized mice contained a higher percentage of the interleukin (IL)-10+ subset, which were able to suppress effector CD4+ T-cells functionally, without any differences between p210-CTB and controls in FoxP3, IL-10, or transforming growth factor beta (TGF-β) mRNA expression in the aorta.20 Furthermore, there was no difference in the numbers of FoxP3+ cells in aortic lesions or CD4+FoxP3+ T-cells in lung mucosa.20 When p210 was delivered subcutaneously via an implanted mini-osmotic pump as a part of mixture of apoB-100 peptides (p210, malondialdehyde-modified-p210 and p240) or alone for 2 weeks, such treatment reduced atherosclerotic lesions in aortic sinuses and also retarded the progression of established atherosclerotic lesions in old female mice.22 Subcutaneous peptide delivery was associated with reduced activation of CD4+ T-cells and increased the CD4+CD25+FoxP3+ subset of T-cells in lymph nodes. Ablation of CD25+ T-cells by CD25-depleting antibody abrogated the atheroprotective effects of subcutaneous infusion of apoB-100 peptides, similar to the study by Wigren and colleagues.21 Taking these reports together, immunization with p210 clearly elicited a CD4+ T-cell response (be it induction of CD4+CD25+ or CD4+IL-10+ T-cells). How such CD4+ T-cell response is elicited or whether these CD4+ T-cells directly mediate the atheroprotective effect of p210 immunization remains unknown.

Regardless of which cellular immune responses were elicited or how and which form of p210 vaccine was delivered, the observed consistent reduction of atherosclerosis after p210 immunization strongly suggests that p210 is a promising candidate antigen for vaccine formulation optimization for potential future human testing.

Other apoB-100-related antigens

Many investigators have also tested additional apoB-100-derived peptides as potential antigens for vaccine formulation. Immunization with an apoB-100 peptide (amino acid residues 688–707) incorporated into a multiantigenic construct with peptidic epitopes from Chlamydophila pneumoniae and HSP60 reduced atherosclerosis accompanied by a reduction of macrophage infiltration and an increase of CD4+FoxP3 T-cells in the plaques.23

Dr Klaus Ley’s group surveyed the murine apoB-100 protein for peptide fragments that were predicted to bind to the mouse MHC-II molecule I-Ab by modeling algorithms. Overall, two peptide fragments, ApoB3501–3516 and ApoB978–993, were identified and were able to reduce atherosclerosis when used to immunize apoE-/- mice, possibly via an IL-10-dependent mechanism.24

Using T-cell hybridomas generated from human apoB-100 transgenic mice immunized with human oxidized LDL (oxLDL), Dr Goran Hansson’s group was able to identify major histocompatibility class (MHC) class II-restricted, ApoB-100-responding CD4+ T-cell hybridomas expressing a single T-cell receptor V beta chain, TRBV31. Immunizing with a TRBV31-derived peptide induced TRBV31 antibodies that blocked T-cell recognition of apoB-100 and significantly reduced atherosclerosis.25 This innovative approach identified a potentially pathogenic CD4+ T-cell population and used antigen-specific humoral immunity to block a proatherogenic cellular immune response, hence confirming the pathogenic role of CD4+ T-cells in atherosclerosis.

Other lipid-related antigens

The complexity of atherosclerotic vascular disease presents the opportunity to target other potential sources of antigens. The search for suitable antigens for use in vaccines to modulate atherosclerosis has expanded to molecules other than LDL or apoB-100.

A natural IgM antibody recognizing the epitopes in oxLDL26,27 and phosphorylcholine (PC) headgroups on the surface of apoptotic cells, and which inhibits uptake of oxLDL and apoptotic cells by macrophages, has been extensively studied.2729 Protection against infection from Streptococcus pneumonia is attributed to anti-PC antibodies.30,31 Active immunization with S. pneumoniae in LDL-R-/- mice to induce anti-PC antibodies resulted in increased oxLDL antibodies, primarily of the IgM isotype, and reduced atherosclerosis.32 The increase in oxLDL-specific IgM is attributed to the cross-reactivity of the phosphorylcholine moiety on oxLDL with S. pneumonia-induced antibodies, suggesting molecular mimicry between S. pneumoniae and oxLDL. This molecular mimicry was investigated further in the context of a vaccine using PC, the reported mimotope. Immunization of apoE-/- mice with PC-keyhole limpet hemocyanin (KLH)-conjugate coupled to unmethylated cytosine-guanine dinucleotides (CpG) oligonucleotides as adjuvant significantly increased IgG and IgM levels against PC and oxLDL, with reduced macrophage oxLDL uptake, and reduced atherosclerosis.33 However, using myocardial infarction or stroke as endpoints, observational cohort studies in humans did not show consistent protective effects of pneumococcal vaccines,3437 leaving the question of whether active immunization against PC will reduce atherosclerosis in humans unanswered.

Cholesteryl ester transfer protein (CETP) is a key enzyme in the high-density lipoprotein (HDL) metabolic pathway. Immunization of rabbits against CETP-induced neutralizing antibodies and markedly increased High density lipoprotein-cholesterol (HDL-C) levels concomitant with reduced atherosclerosis.3840 Nasal immunization of rabbits with a vaccine targeting both CETP and heat shock protein-65 (HSP65) has also been shown to reduce aortic atherosclerosis.41 However, a phase I human trial did not show consistent induction of CETP antibody nor significant changes in CETP function or HDL levels with CETP immunization.42

Rider and colleagues have eluted peptides from murine MHC-II molecules and found these peptides are predominantly fragments of self-proteins. Among these eluted peptides is Ep1 (237–252), an apoE-derived peptide.43 As a part of Ep1, Ep1.B (239–252) was able to reduce early atherosclerosis when administered intravenously.44 The atheroprotective effect of Ep1.B is thought to be due to the induction of plasmacytoid dendritic cells to generate regulatory T-cells, hence the induction of peripheral tolerance in adaptive immune responses toward atherogenesis.45

Vaccines targeting heat shock protein

Heat shock proteins (HSPs) are stress proteins that are highly conserved in all organisms and can be expressed at high levels when cells are exposed to stresses, such as altered pH or oxygen deprivation. HSPs have also been implicated in atherogenesis.4649 However, the effect of immunization with HSPs on atherosclerosis has been inconsistent. Several groups reported that immunization with HSP65 induces atherosclerotic lesions,5052 whereas others reported reduced atherosclerotic lesions.5355 The difference in outcomes could be attributed to the difference in the adjuvant used or the mode of antigen delivery.

HSP-based vaccines delivered via mucosal approach can elicit a down-modulation of immune responses to specific antigens. Intranasal vaccinations using either plasmid DNA encoding HSP65 or whole protein HSP65, or both in phosphate buffered saline (PBS) in rabbits induced HSP65 IgG responses, increased serum IL-10, and reduced interferon (IFN)-γ, and reduced atherosclerosis accompanied by decreased cholesterol levels.55 Sublingual immunization of hypercholesterolemic mice with a recombinant HSP60 from Porphyromonas gingivalis prevented P. gingivalis-accelerated atherosclerosis by inducing an increase of IFN-γ+ or IL-10+Foxp3+ cells in draining lymph nodes and reduction of serum levels of C-reactive protein (CRP), monocyte chemotactic protein 1 (MCP-1) and oxLDL.56 Another group of investigators reported nasal immunization with HSP60 attenuated atherosclerosis in aortic root with the induction of CD4+GARP+ T-regulatory cells (Tregs), Type 1 regulatory T cell (Tr1) cells and CD4+CD25+FoxP3+ Tregs.57

Vaccines against host cell surface, extracellular matrix proteins or plasma proteins

The involvement of certain inflammatory cells in atherosclerotic plaque formation suggested that specific cell surface markers could be potential antigens for immunization. Oral DNA vaccines targeting cell surface proteins thought to contribute to atherosclerosis have been tested experimentally with success. By delivering the antigen via an expression plasmid that encodes the antigen, this strategy transfers the genetic material from the carrier to host phagocytes in the gastrointestinal tract. The phagocytes then express the antigen de novo in the cytosol, and present it on MHC molecules.58 In the reported studies using this approach, constructs were designed to encode CD9959 or vascular endothelial growth factor receptor 2 (VEGFR2)60 carried by live attenuated S. typhimurium and delivered orally. The expressed antigens were presented by MHC-I, which elicited a CD8+ cytolytic T-cell response targeting cells that expressed VEGFR2 or CD99, resulting in reduced atherosclerosis.

LDL retention in the arterial wall by extracellular matrix (ECM) is an early step in the development of atherosclerotic lesions. Fibronectin is an ECM protein found in plaques. Immunization with fibronectin formulated with alum as the adjuvant significantly reduced atherosclerosis in apoE-/- mice, and was associated with increased Th2-type antibody production and increased regulatory T-cells.61 Interestingly, plasma cholesterol was significantly reduced in the immunized mice, suggesting an interaction between immune responses to ECM proteins and cholesterol metabolism.

β2-glycoprotein I (β2-GPI) is a glycosylated plasma protein that has been implicated to play a role in atherogenesis. Immunization with β2-GPI attenuated the development of early atherosclerosis, presumably by inducing tolerance against β2-GPI.62 Immunization with peptides from the N-terminus of the C5a receptor also reduced early atherosclerotic lesion formation possibly via induction of regulatory T-cell response.63 Pro-inflammatory cytokine IL-1α is another potential target for vaccine development. Immunization with full-length, native IL-1α chemically conjugated to virus-like particles reduced inflammatory response in the plaques and atherosclerosis progression in aorta and aortic root.64

PCSK9 vaccines

Proprotein convertase subtilisin/kexin type 9 (PCSK9) secreted by the liver negatively regulates the LDL-receptor (LDL-R) by binding to LDL-R and blocking the recycling of the receptor to the cell surface, thus decreasing the uptake of LDL particles and increasing the circulating level of LDL-C. The loss of function of PCSK9 mutation in humans results in life-long hypocholesterolemia and lower incidence of cardiovascular events.65 Antibodies against PCSK9 have been developed and successfully reduced LDL-C levels in conjunction with use of statin in clinical trials.66,67 Evolocumab (Amgen, USA), alirocumab (Sanofi/Regeneron, France/USA) and bococizumab (Pfizer/Rinat, USA) are now undergoing phase III clinical trial testing to determine their clinical efficacy to reduce cardiovascular events. However, the biggest disadvantage of using a PCSK9 antibody as a treatment strategy is the need for repeated injections and its associated high cost, potentially limiting the wide clinical use of the PCSK9 antibody. Thus investigators have been testing the notion of active immunization against PCSK9 to achieve similar but long-term biological effects to the use of PCSK9 antibody. In preclinical experiments, PCSK9 peptide-based vaccines have been shown to elicit a PCSK9-specific antibody with a significant reduction of LDL-C.68,69 If these vaccines can be further demonstrated to be well tolerated and effective for clinical application, active immunization against PCSK9 will be an attractive alternative to the PCSK9 antibody.

Implications and clinical perspectives

The idea of developing vaccination strategies to modulate the highly prevalent atherosclerotic cardiovascular disease is exciting and daunting but still in its infancy. In this review, we discussed the experimental evidence and efficacy of reducing atherosclerosis by vaccination using many different antigens tested in preclinical models. Before translating the aforementioned promising preclinical observations into the clinical arena, we need to answer many challenging questions while designing proper clinical studies. We believe there is currently no lack of suitable antigens to be tested in vaccine formulation for clinical testing. Numerous clinical trials have established the causative role of LDL in atherogenesis, which makes LDL and its apoB-100 reasonable and logical initial targets for vaccination. This is also supported by preclinical studies using LDL or apoB-100 peptides as candidate antigens in vaccine formulation (Table 1). The challenges reside in the following areas: choice of formulation and route of delivery, vaccine safety and stability, schedule and durability of immunization, proper selection of patient populations for testing, and determination and monitoring of efficacy endpoints in clinical studies. Potential side effects of immunization such as undesirable immune activation, whether they are specifically related to atherogenesis or not, are the additional challenges that need to be addressed in early safety trials. This is going to be a costly and long journey. We have the vision of moving this idea of vaccination to reduce atherosclerosis into clinical testing, but this goal cannot be achieved without academic investigators, professional societies, government agencies, funding organizations, and the pharmaceutical industries working together to initiate this long journey. With all of these challenges in mind, we are cautiously optimistic about the potential for future clinical application.

Table 1.

Summary of published reports utilizing LDL, modified LDL or apoB-100 related peptides as antigens for immunization.

LDL or its modified form
Animal Antigens Adjuvant Immunization route Effect on atherosclerosis Reference
LDL-R-/- rabbits MDA-LDL Freund’s complete followed by incomplete adjuvant Subcutaneous followed by intramuscular Reduced (aorta) Palinski et al.10
NZW rabbits on high cholesterol diet Native LDL or Cuox-LDL AdjuPrime (carbohydrate polymer) Subcutaneous Reduced (aorta) Ameli et al.6
LDL-R-/- mice Native LDL or MDA-LDL Freund’s complete followed by incomplete adjuvant Subcutaneous followed by intraperitoneal Reduced (aortic sinus) Freigang et al.8
ApoE-/- mice MDA-LDL Freund’s complete followed by incomplete adjuvant Subcutaneous Reduced (aortic sinus) George et al.9
ApoE-/- mice Plaque homogenate or MDA-LDL Freund’s complete followed by incomplete adjuvant Foot pad injection Reduced (aortic sinus) Zhou et al.12
ApoE-/- mice Native LDL IL-12 Subcutaneous Reduced (aortic sinus) Chyu et al.7
ApoE-/- or apoE/CD4 double knockout mice MDA-LDL Freund’s complete followed by incomplete adjuvant Subcutaneous Reduced (aortic sinus) Zhou et al.13
LDL-R-/- mice Cuox-LDL Dendritic cells Intravenous delivery of oxLDL pulsed dendritic cells Reduced (accelerated carotid atherosclerosis induced by pericarotid collar) Habets, 201070
ApoE-/- mice Cuox-LDL None Nasal delivery Reduced (aortic sinus and aorta) Zhang et al.52
LDL-R-/- and apoE-/- mice Cuox-LDL or AGE-LDL Alum (Pierce) Subcutaneous Reduced (aortic sinus and aorta) Zhu et al.14
apoB-100 peptide
Animal Antigens Adjuvant Immunization route Effect on atherosclerosis Reference
ApoE-/- mice Mixture of p143 and p210 Alum (Pierce) NA Reduced (descending aorta) Fredrikson, 2003b
ApoE-/- mice MDA-modified p45 or p74 Alum (Pierce) NA Reduced (descending aorta) Fredrikson, 200571
ApoE-/- mice p2 Alum (Pierce) Subcutaneous followed by intraperitoneal Reduced (aorta) Chyu et al.16
LDL-R(-/-)/human apoB-100 transgenic mice p210 Alum (Pierce) NA Reduced (descending aorta) Fredrikson et al.19
ApoE-/- mice p210 CTB (p210-CTB fusion protein) Intranasal Reduced (aortic sinus) Klingenberg et al.20
ApoE-/- mice p210 Alum (Pierce) Subcutaneous Reduced (descending aorta) Wigren et al.21
ApoE-/- mice Mixture of p210, MDA-p210 and p240 or p210 only No adjuvant Continuous subcutaneously delivery Reduced (aortic sinus) Herbin et al.22
ApoE-/- mice p210 Alum (Pierce) Subcutaneous Reduced (aorta) Chyu et al.18
ApoE-/- mice ApoB3501–3516 or ApoB978–993 Freund’s complete followed by incomplete adjuvant Subcutaneous followed by intraperitoneal Reduced (aortic sinus and aorta) Tse et al.24

CTB, cholera toxin B; Cuox, copper oxidized; IL, interleukin; LDL, low-density lipoprotein; LDL-R, LDL receptor; MDA, malondialdehyde; NA, not applicable; NZW, New Zealand White; oxLDL, oxidized LDL; SQ, subcutaneously.

Acknowledgments

Dr Chyu and Dr Dimayuga contributed equally to this paper.

Footnotes

Funding: Funding for this study was provided by the Eisner Foundation, USA, The Heart Foundation, USA, the Spielberg Cardiovascular Research Fund, USA, Petersen Foundation, USA, Annenberg Foundation, USA, Steinberg Foundation and Cardiovax, Inc, USA.

Conflict of interest statement: Dr Chyu and Dr Shah are co-inventors of the apoB-100-based peptide vaccine. Patent rights are assigned to Cedars-Sinai Medical Center, CA, USA.

Contributor Information

Kuang-Yuh Chyu, Oppenheimer Atherosclerosis Research Center, Division of Cardiology, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.

Paul C. Dimayuga, Oppenheimer Atherosclerosis Research Center, Division of Cardiology, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA

Prediman K. Shah, Cedars-Sinai Medical Center, 127 South San Vicente Blvd., Suite A-3307, Los Angeles, CA 90048, USA.

References

  • 1. Ammirati E, Moroni F, Magnoni M, et al. The role of T and B cells in human atherosclerosis and atherothrombosis. Clin Exp Immunol 2015; 179: 173–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Libby P, Lichtman AH, Hansson GK. Immune effector mechanisms implicated in atherosclerosis: from mice to humans. Immunity 2013; 38: 1092–1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Libby P, Hansson GK. Inflammation and immunity in diseases of the arterial tree: players and layers. Circ Res 2015; 116: 307–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Tsiantoulas D, Diehl CJ, Witztum JL, et al. B cells and humoral immunity in atherosclerosis. Circ Res 2014; 114: 1743–1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Ketelhuth DF, Hansson GK. Adaptive response of T and B cells in atherosclerosis. Circ Res 2016; 118: 668–678. [DOI] [PubMed] [Google Scholar]
  • 6. Ameli S, Hultgardh-Nilsson A, Regnstrom J, et al. Effect of immunization with homologous LDL and oxidized LDL on early atherosclerosis in hypercholesterolemic rabbits. Arterioscler Thromb Vasc Biol 1996; 16: 1074–1079. [DOI] [PubMed] [Google Scholar]
  • 7. Chyu KY, Reyes OS, Zhao X, et al. Timing affects the efficacy of LDL immunization on atherosclerotic lesions in apoE (-/-) mice. Atherosclerosis 2004; 176: 27–35. [DOI] [PubMed] [Google Scholar]
  • 8. Freigang S, Horkko S, Miller E, et al. Immunization of LDL receptor-deficient mice with homologous malondialdehyde-modified and native LDL reduces progression of atherosclerosis by mechanisms other than induction of high titers of antibodies to oxidative neoepitopes. Arterioscler Thromb Vasc Biol 1998; 18: 1972–1982. [DOI] [PubMed] [Google Scholar]
  • 9. George J, Afek A, Gilburd B, et al. Hyperimmunization of apo-E-deficient mice with homologous malondialdehyde low-density lipoprotein suppresses early atherogenesis. Atherosclerosis 1998; 138: 147–152. [DOI] [PubMed] [Google Scholar]
  • 10. 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 U S A 1995; 92: 821–825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Zhong Y, Wang X, Ji Q, et al. CD4+LAP + and CD4 +CD25 +Foxp3 + regulatory T cells induced by nasal oxidized low-density lipoprotein suppress effector T cells response and attenuate atherosclerosis in apoE-/- mice. J Clin Immunol 2012; 32: 1104–1117. [DOI] [PubMed] [Google Scholar]
  • 12. Zhou X, Caligiuri G, Hamsten A, et al. LDL immunization induces T-cell-dependent antibody formation and protection against atherosclerosis. Arterioscler Thromb Vasc Biol 2001; 21: 108–114. [DOI] [PubMed] [Google Scholar]
  • 13. Zhou X, Robertson AK, Rudling M, et al. Lesion development and response to immunization reveal a complex role for CD4 in atherosclerosis. Circ Res 2005; 96: 427–434. [DOI] [PubMed] [Google Scholar]
  • 14. Zhu L, He Z, Wu F, et al. Immunization with advanced glycation end products modified low density lipoprotein inhibits atherosclerosis progression in diabetic apoE and LDLR null mice. Cardiovasc Diabetol 2014; 13: 151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Fredrikson GN, Hedblad B, Berglund G, et al. Identification of immune responses against aldehyde-modified peptide sequences in apoB associated with cardiovascular disease. Arterioscler Thromb Vasc Biol 2003; 23: 872–878. [DOI] [PubMed] [Google Scholar]
  • 16. Chyu KY, Zhao X, Reyes OS, et al. Immunization using an apoB-100 related epitope reduces atherosclerosis and plaque inflammation in hypercholesterolemic apoE (-/-) mice. Biochem Biophys Res Commun 2005; 338: 1982–1989. [DOI] [PubMed] [Google Scholar]
  • 17. Fredrikson GN, Soderberg I, Lindholm M, et al. Inhibition of atherosclerosis in apoE-null mice by immunization with apoB-100 peptide sequences. Arterioscler Thromb Vasc Biol 2003; 23: 879–884. [DOI] [PubMed] [Google Scholar]
  • 18. Chyu KY, Zhao X, Dimayuga PC, et al. CD8+ T cells mediate the athero-protective effect of immunization with an apoB-100 peptide. PLoS ONE 2012; 7: e30780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Fredrikson GN, Bjorkbacka H, Soderberg I, et al. Treatment with apoB peptide vaccines inhibits atherosclerosis in human apoB-100 transgenic mice without inducing an increase in peptide-specific antibodies. J Intern Med 2008; 264: 563–570. [DOI] [PubMed] [Google Scholar]
  • 20. Klingenberg R, Lebens M, Hermansson A, et al. Intranasal immunization with an apolipoprotein B-100 fusion protein induces antigen-specific regulatory T cells and reduces atherosclerosis. Arterioscler Thromb Vasc Biol 2010; 30: 946–952. [DOI] [PubMed] [Google Scholar]
  • 21. Wigren M, Kolbus D, Duner P, et al. Evidence for a role of regulatory T cells in mediating the atheroprotective effect of apolipoprotein B peptide vaccine. J Intern Med 2010; 269: 546–556. [DOI] [PubMed] [Google Scholar]
  • 22. Herbin O, it-Oufella H, Yu W, et al. Regulatory T-cell response to apolipoprotein B100-derived peptides reduces the development and progression of atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2012; 32: 605–612. [DOI] [PubMed] [Google Scholar]
  • 23. Lu X, Xia M, Endresz V, et al. Impact of multiple antigenic epitopes from apoB100, hHSP60 and Chlamydophila pneumoniae on atherosclerotic lesion development in apoB(tm2Sgy)Ldlr(tm1Her)J mice. Atherosclerosis 2012; 225: 56–68. [DOI] [PubMed] [Google Scholar]
  • 24. Tse K, Gonen A, Sidney J, et al. Atheroprotective vaccination with MHC-II restricted peptides from apoB-100. Front Immunol 2013; 4: 493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Hermansson A, Ketelhuth DF, Strodthoff D, et al. Inhibition of T cell response to native low-density lipoprotein reduces atherosclerosis. J Exp Med 2010; 207: 1081–1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Friedman P, Horkko S, Steinberg D, et al. Correlation of antiphospholipid antibody recognition with the structure of synthetic oxidized phospholipids: importance of Schiff base formation and aldol condensation. J Biol Chem 2002; 277: 7010–7020. [DOI] [PubMed] [Google Scholar]
  • 27. Horkko S, Bird DA, Miller E, et al. Monoclonal autoantibodies specific for oxidized phospholipids or oxidized phospholipid-protein adducts inhibit macrophage uptake of oxidized low-density lipoproteins. J Clin Invest 1999; 103: 117–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Chang MK, Bergmark C, Laurila A, et al. Monoclonal antibodies against oxidized low-density lipoprotein bind to apoptotic cells and inhibit their phagocytosis by elicited macrophages: evidence that oxidation-specific epitopes mediate macrophage recognition. Proc Natl Acad Sci USA 1999; 96: 6353–6358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Shaw PX, Horkko S, Chang MK, et al. Natural antibodies with the T15 idiotype may act in atherosclerosis, apoptotic clearance, and protective immunity. J Clin Invest 2000; 105: 1731–1740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Briles DE, Forman C, Hudak S, et al. Anti-phosphorylcholine antibodies of the T15 idiotype are optimally protective against Streptococcus pneumoniae. J Exp Med 1982; 156: 1177–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Yother J, Forman C, Gray BM, et al. Protection of mice from infection with Streptococcus pneumoniae by anti-phosphocholine antibody. Infect Immun 1982; 36: 184–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Binder CJ, Horkko S, Dewan A, et al. Pneumococcal vaccination decreases atherosclerotic lesion formation: molecular mimicry between Streptococcus pneumoniae and oxidized LDL. Nat Med 2003; 9: 736–743. [DOI] [PubMed] [Google Scholar]
  • 33. Caligiuri G, Khallou-Laschet J, Vandaele M, et al. Phosphorylcholine-targeting immunization reduces atherosclerosis. J Am Coll Cardiol 2007; 50: 540–546. [DOI] [PubMed] [Google Scholar]
  • 34. Hung IF, Leung AY, Chu DW, et al. Prevention of acute myocardial infarction and stroke among elderly persons by dual pneumococcal and influenza vaccination: a prospective cohort study. Clin Infect Dis 2010; 51: 1007–1016. [DOI] [PubMed] [Google Scholar]
  • 35. Lamontagne F, Garant MP, Carvalho JC, et al. Pneumococcal vaccination and risk of myocardial infarction. Can Med Assoc J 2008; 179: 773–777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Siriwardena AN, Gwini SM, Coupland CA. Influenza vaccination, pneumococcal vaccination and risk of acute myocardial infarction: matched case-control study. Can Med Assoc J 2010; 182: 1617–1623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Tseng HF, Slezak JM, Quinn VP, et al. Pneumococcal vaccination and risk of acute myocardial infarction and stroke in men. J Am Med Assoc 2010; 303: 1699–1706. [DOI] [PubMed] [Google Scholar]
  • 38. Gaofu Q, Jun L, Xin Y, et al. Vaccinating rabbits with a cholesteryl ester transfer protein (CETP) B-Cell epitope carried by heat shock protein-65 (HSP65) for inducing anti-CETP antibodies and reducing aortic lesions in vivo. J Cardiovasc Pharmacol 2005; 45: 591–598. [DOI] [PubMed] [Google Scholar]
  • 39. Mao D, Kai G, Gaofu Q, et al. Intramuscular immunization with a DNA vaccine encoding a 26-amino acid CETP epitope displayed by HBc protein and containing CpG DNA inhibits atherosclerosis in a rabbit model of atherosclerosis. Vaccine 2006; 24: 4942–4950. [DOI] [PubMed] [Google Scholar]
  • 40. Rittershaus CW, Miller DP, Thomas LJ, et al. Vaccine-induced antibodies inhibit CETP activity in vivo and reduce aortic lesions in a rabbit model of atherosclerosis. Arterioscler Thromb Vasc Biol 2000; 20: 2106–2112. [DOI] [PubMed] [Google Scholar]
  • 41. Jun L, Jie L, Dongping Y, et al. Effects of nasal immunization of multi-target preventive vaccines on atherosclerosis. Vaccine 2012; 30: 1029–1037. [DOI] [PubMed] [Google Scholar]
  • 42. Davidson MH, Maki K, Umporowicz D, et al. The safety and immunogenicity of a CETP vaccine in healthy adults. Atherosclerosis 2003; 169: 113–120. [DOI] [PubMed] [Google Scholar]
  • 43. Rider BJ, Fraga E, Yu Q, et al. Immune responses to self peptides naturally presented by murine class II major histocompatibility complex molecules. Mol Immunol 1996; 33: 625–633. [DOI] [PubMed] [Google Scholar]
  • 44. Bocksch L, Rider BJ, Stephens T, et al. C-terminal apolipoprotein E-derived peptide, Ep1.B, displays anti-atherogenic activity. Atherosclerosis 2007; 194: 116–124. [DOI] [PubMed] [Google Scholar]
  • 45. Bellemore SM, Nikoopour E, Au BC, et al. Anti-atherogenic peptide Ep1.B derived from apolipoprotein E induces tolerogenic plasmacytoid dendritic cells. Clin Exp Immunol 2014; 177: 732–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Almanzar G, Ollinger R, Leuenberger J, et al. Autoreactive HSP60 epitope-specific T-cells in early human atherosclerotic lesions. J Autoimmun 2012; 39: 441–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Grundtman C, Kreutmayer SB, Almanzar G, et al. Heat shock protein 60 and immune inflammatory responses in atherosclerosis. Arterioscler Thromb Vasc Biol 2011; 31: 960–968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Grundtman C, Wick G. The autoimmune concept of atherosclerosis. Curr Opin Lipidol 2011; 22: 327–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Kilic A, Mandal K. Heat shock proteins: pathogenic role in atherosclerosis and potential therapeutic implications. Autoimmune Dis 2012; 2012: 502813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. George J, Shoenfeld Y, Afek A, et al. Enhanced fatty streak formation in C57BL/6J mice by immunization with heat shock protein-65. Arterioscler Thromb Vasc Biol 1999; 19: 505–510. [DOI] [PubMed] [Google Scholar]
  • 51. Xu Q, Dietrich H, Steiner HJ, et al. Induction of arteriosclerosis in normocholesterolemic rabbits by immunization with heat shock protein 65. Arterioscler Thromb 1992; 12: 789–799. [DOI] [PubMed] [Google Scholar]
  • 52. Zhang Y, Xiong Q, Hu X, et al. A novel atherogenic epitope from Mycobacterium tuberculosis heat shock protein 65 enhances atherosclerosis in rabbit and LDL receptor-deficient mice. Heart Vessels 2012; 27: 411–418. [DOI] [PubMed] [Google Scholar]
  • 53. Grundtman C, Jakic B, Buszko M, et al. Mycobacterial heat shock protein 65 (mbHSP65)-induced atherosclerosis: preventive oral tolerization and definition of atheroprotective and atherogenic mbHSP65 peptides. Atherosclerosis 2015; 242: 303–310. [DOI] [PubMed] [Google Scholar]
  • 54. Klingenberg R, Ketelhuth DF, Strodthoff D, et al. Subcutaneous immunization with heat shock protein-65 reduces atherosclerosis in apoE(-)/(-) mice. Immunobiology 2012; 217: 540–547. [DOI] [PubMed] [Google Scholar]
  • 55. Long J, Lin J, Yang X, et al. Nasal immunization with different forms of heat shock protein-65 reduced high-cholesterol-diet-driven rabbit atherosclerosis. Int Immunopharmacol 2012; 13: 82–87. [DOI] [PubMed] [Google Scholar]
  • 56. Hagiwara M, Kurita-Ochiai T, Kobayashi R, et al. Sublingual vaccine with GroEL attenuates atherosclerosis. J Dent Res 2014; 93: 382–387. [DOI] [PubMed] [Google Scholar]
  • 57. Zhong Y, Tang H, Wang X, et al. Intranasal immunization with heat shock protein 60 induces CD4(+) CD25(+) GARP(+) and type 1 regulatory T cells and inhibits early atherosclerosis. Clin Exp Immunol 2016; 183: 452–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Darji A, Guzman CA, Gerstel B, et al. Oral somatic transgene vaccination using attenuated S. typhimurium. Cell 1997; 91: 765–775. [DOI] [PubMed] [Google Scholar]
  • 59. van Wanrooij EJ, De VP, Bixel MG, et al. Vaccination against CD99 inhibits atherogenesis in low-density lipoprotein receptor-deficient mice. Cardiovasc Res 2008; 78: 590–596. [DOI] [PubMed] [Google Scholar]
  • 60. Hauer AD, van Puijvelde GH, Peterse N, et al. Vaccination against VEGFR2 attenuates initiation and progression of atherosclerosis. Arterioscler Thromb Vasc Biol 2007; 27: 2050–2057. [DOI] [PubMed] [Google Scholar]
  • 61. Duner P, To F, Beckmann K, et al. Immunization of apoE-/- mice with aldehyde-modified fibronectin inhibits the development of atherosclerosis. Cardiovasc Res 2011; 91: 528–536. [DOI] [PubMed] [Google Scholar]
  • 62. De HJ, Esparza L, Bleda S, et al. Attenuation of early atherosclerotic lesions by immunotolerance with beta2 glycoprotein I and the immunomodulatory effectors interleukin 2 and 10 in a murine model. J Vasc Surg 2015; 62: 1625–1631. [DOI] [PubMed] [Google Scholar]
  • 63. Lu X, Xia M, Endresz V, et al. Immunization with a combination of 2 peptides derived from the C5a receptor significantly reduces early atherosclerotic lesion in Ldlr(tm1Her) apoB(tm2Sgy) J mice. Arterioscler Thromb Vasc Biol 2012; 32: 2358–2371. [DOI] [PubMed] [Google Scholar]
  • 64. Tissot AC, Spohn G, Jennings GT, et al. A VLP-based vaccine against interleukin-1alpha protects mice from atherosclerosis. Eur J Immunol 2013; 43: 716–722. [DOI] [PubMed] [Google Scholar]
  • 65. Cohen JC, Boerwinkle E, Mosley TH, Jr, et al. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med 2006; 354: 1264–1272. [DOI] [PubMed] [Google Scholar]
  • 66. Farnier M. Future lipid-altering therapeutic options targeting residual cardiovascular risk. Curr Cardiol Rep 2016; 18: 65. [DOI] [PubMed] [Google Scholar]
  • 67. Feinstein MJ, Lloyd-Jones DM. Monoclonal antibodies for lipid management. Curr Atheroscler Rep 2016; 18: 39. [DOI] [PubMed] [Google Scholar]
  • 68. Crossey E, Amar MJ, Sampson M, et al. A cholesterol-lowering VLP vaccine that targets PCSK9. Vaccine 2015; 33: 5747–5755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Galabova G, Brunner S, Winsauer G, et al. Peptide-based anti-PCSK9 vaccines - an approach for long-term LDLc management. PLoS One 2014; 9: e114469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Habets KL, van Puijvelde GH, et al. Vaccination using oxidized low-density lipoprotein-pulsed dendritic cells reduces atherosclerosis in LDL receptor-deficient mice. Cardiovasc Res 2010; 85: 622–630. [DOI] [PubMed] [Google Scholar]
  • 71. Fredrikson GN, Anderson L, et al. . Atheroprotective immunization with MDA-modified apo B-100 peptide sequences is associated with activation of Th2 specific antibody expression. Autoimmunity 2005; 38: 171–179. [DOI] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Vaccines are provided here courtesy of SAGE Publications

RESOURCES