Review

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

The duality of chemokines in heart failure Expert Rev. Clin. Immunol. 11(4), 523–536 (2015)

Andrew A Jarrah and Sima T Tarzami* Department of Medicine, Division of Cardiovascular Research Center, Mount Sinai School of Medicine, 1 Gustave L Levy Place, Box 1030, New York, NY 10029, USA *Author for correspondence: Tel.: +1 212 241 8228 Fax: +1 212 241 4080 [email protected]

The failing human heart is a bustling network of intra- and inter-cellular signals and related processes attempting to coordinate a repair mechanism for the injured or diseased myocardium. While our understanding of signaling by mode of cytokines is well understood on a systemic level, we are only now coming to elucidate the role of cytokines in cardiac self-regulation. An increasing number of studies are showing now that cardiomyocytes themselves have not only the ability but also the mandate to produce signals, and play direct roles in how these signals are interpreted. One of the families of cytokines employed by distressed cardiac tissue are chemokines. By regulating the movement of pro-inflammatory cell types to sites of injury, we see now how the myocardium responds to stress. Herein we review the participation of these inflammatory mediators and explore the delicate balance between their protective roles and damaging functions. KEYWORDS: anti-cytokine therapies . chemokines . clinical trials . cytokines . heart failure . stem cell-based cytokine therapies

Heart disease is set to break 100 years as the number one killer in the USA and a major cause of death in the industrialized world by the end of the current decade. Each year, roughly three-quarters of a million Americans die from cardiovascular disease [1]. We can only expect a significant increase in the number of patients requiring cardiac care as the American population begins to age [1]. The need to not only mitigate heart disease, but develop penetrating and efficient therapies is an urgent one, and it relies heavily on our ability to effectively seek out targets and manipulate them. Cytokines & heart failure

The hypothesis that proinflammatory cytokines induce left ventricular dysfunction was first deduced from animal [2] and human studies of sepsis [3]. These hypotheses put forward that sepsis-associated cardiac dysfunction was caused by circulating myocardial depressant substances, later identified, in large part, to be inflammatory cytokines (including TNF-1a, IL-1b, IL-6, IL-2 and IFN-g) [4–6]. These ideas were supported from numerous animal [7–9] and in vitro studies [2,4,10]. Cytokine overexpression replicates a heart failure phenotype, and blocking the respective cytokine [11] or its respective receptors ameliorates the phenotype [12–15]. Further correlative studies demonstrate a positive informahealthcare.com

10.1586/1744666X.2015.1024658

relationship between cytokine levels and progressively worsening heart failure [13,16,17]. The substantive body of experimental and clinical observations as well as the obvious translational nature of the animal studies led to a series of multicenter clinical trials (TABLE 1) that used ‘targeted’ approaches to neutralize cytokines, such as TNF-a, in patients with moderate to advanced heart failure. However, these targeted approaches resulted in worsening heart failure, thereby raising a number of important questions about what role, if any, proinflammatory cytokines play in the pathogenesis of heart failure. Chemokines have also been implicated in the pathogenesis of various forms of heart failure where elevated levels have been found in patients with failing hearts [18,19]. Using animal studies, it has been demonstrated that chemokines play important regulatory roles in a myriad of physiological functions such as inflammation, autoimmunity, lymphoid organogenesis, vascular angiogenesis and cell survival [20–24]. Chemokines

Chemokines are a class of cytokines that have chemoattractant properties. They are small glycoproteins that signal through septahelical G protein-coupled receptors, a large and functionally diverse superfamily of cell surface

 2015 Informa UK Ltd

ISSN 1744-666X

523

Review

Jarrah & Tarzami

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

Table 1. Anticytokine therapies: TNF-a inhibition. Drug

TNF-a antagonists

Mechanism of action

Clinical trials

Outcome

Ref.

Etanercept

A soluble p75 TNF-a receptor fusion protein

Binds and inactivates TNF-a

RENAISSANCE (USA) study for heart failure

Failed to show any improvements

[11,172]

Etanercept

A soluble p75 TNF-a receptor fusion protein

Binds and inactivates TNF-a

RECOVER study for heart failure

Failed to show any improvements

[169,170,172]

Etanercept

A soluble p75 TNF-a receptor fusion protein

Binds and inactivates TNF-a

RENEWAL update on (RENAISSANCE + RECOVER) for heart failure

Failed to show any improvements

[171,172]

Infliximab

A chimeric (mouse/human) anti-TNF-a Ab

Neutralizes soluble and membrane-bound TNF-a

ATTACH study for heart failure

Failed to show any improvements

[168]

Adalimumab

A fully human monoclonal anti-TNF-a Ab

Neutralizes soluble and membrane-bound TNF-a

PREMIER study for rheumatoid arthritis

Showed benefit in combination with methotrexate

[173]

Certolizumab pegol

An antigen-binding fragment (Fab’) of a humanized mAb coupled to polyethylene glycol

Neutralizes soluble and membrane-bound TNF-a

RAPID1 & 2 study for rheumatoid arthritis

Showed benefit

[174]

Certolizumab pegol

An antigen-binding fragment (Fab’) of a humanized mAb coupled to polyethylene glycol

Neutralizes soluble and membrane-bound TNF-a

FAST4WARD study for rheumatoid arthritis

Showed benefit

[175]

Golimumab

A human anti-TNF-a mAb

Neutralizes soluble and membrane-bound TNF-a

PURSUITS study for colitis

Showed benefit

[176]

mAb: Monoclonal antibody.

receptors mediating a large number of cellular responses to extracellular stimuli [25–27]. There are significant structural and regulatory differences in the functional mechanisms of cytokine receptors such as the TNFR and IL-2R as compared to chemokine receptors. First, cytokines are proinflammatory molecules, induced in infection and immune activation whereas chemokine receptors are expressed either inducibly (inflammatory) or constitutively [28]. Chemokines have been classified into four main subfamilies depending on the spacing of their first two cysteine residues: CXC (or a-chemokines), CC (or b-chemokines), CX3C and XC. All of these proteins exert their effects by binding with G protein-coupled receptors called chemokine receptors that are found on the surfaces of their target cells. In addition to their chemotactic and cell homing properties, both chemokines and their receptors are expressed and have entirely distinct biological functions on many cell types in the body including fibroblasts, endothelial cells, smooth muscle cells, cardiomyocytes, neurons and so on [29–32]. While much of their form and function has been uncovered on the universal level, there are still unresolved questions when put into the context of the heart. In the disease state, the characterization of these receptors and their ligands is ongoing, with the role of chemokines being understood only recently. Chemokines & heart failure

Observational studies have demonstrated the presence of certain chemokines within the dysfunctional heart [33–36]. The work of our laboratory and others has further elucidated chemokine 524

receptors to be members of the CXC and C-C-chemokines [23,37,38]. The pathogenesis of dysfunction stemmed from a variety of etiologies including myocarditis [39,40], as well as dilated and ischemic cardiomyopathies [19,33,41]. In explanted hearts from 10 patients with end-stage heart failure, using RNase protection assays and immunohistochemistry, Dama˚s et al. demonstrated particularly high mRNA levels of CCL2 [37]. A transgenic approach overexpressing CCL2 resulted in a cardiomyopathic phenotype [42]. Conversely, treatment with 7ND, an N-terminal deletion mutant of the human CCL2 gene, improved survival, attenuated left ventricular dilatation and decreased contractile dysfunction compared with controls in a murine ischemic cardiomyopathy model [43]. None of these studies, however, have postulated potential mechanisms for chemokine effects on myocardial function other than to suggest that the infiltration of lymphocytes and monocytes, along with concomitant cytokine production [43], into the failing myocardium may lead to reversible or irreversible damage of the cardiac muscle [20,44]. The duality of chemokines has at times been confounding, yet the ability of the same axis to play opposite roles suggests that the variability in these signals depends on the injury or disease environment in which they are observed in addition to the cell types that are expressed in. By method of example, we take IL-8 or CXCL8, one of the first chemokines implicated in a cardiac disease state, as a quick study. In the setting of ischemic reperfusion (I/R) injury, studies have illustrated that CXCL8 is a major player responsible for the recruitment of neutrophils, which resulted in greater myocardial injury [20]. Expert Rev. Clin. Immunol. 11(4), (2015)

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

Chemokines in heart failure

Conversely however, our group has shown in vitro that macrophage inflammatory protein-2 (MIP-2 or CXCL2), one of the many murine IL-8 homologues, promoted cell survival by reducing apoptosis [23,24]. The dual nature of these signaling proteins is not segregated to a particular subset; take Regulated on Activation Normal T cell Expressed and Secreted (RANTES or CCL5) and CCL2, which have been shown to play a malignant role in the dysfunction of the myocardium [43,45,46]. However, in experimental animal models of vascular injury, CCL5 exerts proangiogenic effects in vivo and in vitro [47]. Our group and others have also demonstrated that CCL2 is proangiogenic and safeguards cardiomyocytes from hypoxia-induced cell death [22,48], yet those same cardioprotective effects are diminished in the setting of I/R injury where inflammatory cell recruitment was increased [38]. Stromal-cell derived factor-1 (SDF-1 or CXCL12) is yet another prime illustration of the ‘double-edged’ role of cardiac chemokines. In a scenario post-myocardial infarction (MI), CXCL12 has been shown to recruit stem cells and promote neovascularization. Yet Liehn et al. have recently shown that the selective inhibition of CXCL12’s receptor CXCR4 by the antagonist AMD3100 reduced scar formation and enhanced cardiac contractility [49]. The juxtaposition of these findings emphasizes the complexity of these chemokines in disease supporting the notion that we cannot simply separate the various players into the good and the bad, protective or maladaptive. We hope to shed some light on the divisions in this review as we characterize the diverse roles that chemokines play in heart failure. Because we cannot segregate the effects of these chemokines on the heart from other body systems, we will very generally mention the effects of chemokines in a systemic context. Additionally, we will outline the challenges of chemokine/cytokine-based immunotherapies and further discuss their dual function in relation to development of efficient treatment strategies for heart disease. CCL2 (MCP-1)

The CCL2/MCP-1 family is a subfamily of the CC chemokines and comprised of four members [50]. CCL2 negotiates a response in the cell by interacting with its main receptor, CCR2. CCR2 and CCR4 are two cell surface receptors that bind CCL2 [51]. CCL2 is located on 17q11.2 in humans and is the gene product of the JE gene in mice [50,52]. The immature peptide is composed of 99 amino acid residues, with a 23-amino acid signal sequence being cleaved to give the mature protein at 76 amino acids in length [50]. CCL2 is both a potent monocyte and activated T-cell chemoattractant as well as a primary activator for macrophages [53,54]. CCL2 is primarily secreted by monocytes, macrophages and dendritic cells. Apart from known cell types, CCL2 is also expressed in a larger panel of other cell types including osteoblasts, neurons, astrocytes, microglia, smooth muscle cells, fibroblasts and cardiomyocytes [23,32,55–57]. Given the fact that these cell types also express CCR2 (a CCL2 receptor) on the cell surface, these cells can both generate and respond to inflammatory responses, in either an autocrine or a paracrine manner. informahealthcare.com

Review

Published findings have demonstrated the induction of both CCL2 and its receptor CCR2 and implicated them in an assortment of diseases characterized by monocytic infiltrates, such as psoriasis, rheumatoid arthritis and atherosclerosis [58]. There has been a tremendous amount of evidence mounting against CCL2 as one of the main inflammatory cytokines involved in the failure of the heart. Several clinical studies report CCL2 upregulation in patients with diminished cardiac activity and with the infiltration of mononuclear cells marked as the key culprit for cardiac disease development and progression [19,37]. While we are careful not to peg maladaptive to any chemokine in our discussion, CCL2 hyperactivity has been observed in the context of various diseases and injuries. In one study, researchers identified significantly elevated CCL2 levels in the sera of patients 24 h post-MI [59]. The proinflammatory role of CCL2 in congestive heart failure has been well documented [33,60], with several studies concluding that chemokine levels were inversely correlated with left-ventricular ejection fraction, a main determinant of cardiovascular well-being [19]. Additionally, we see clear-cut roles in the pathogenesis of atherosclerosis [61,62], cardiomyopathy [39,63] and myocardial ischemia [64]. However, in addition to pathological effects brought about by the presence of potentially damaging inflammatory cells, that is, monocytes and macrophages, ‘observations suggest that CCL2 was also involved in the repair process.’ CCL2 is shown to act directly on endothelial cells and induce angiogenesis through the induction of VEGF [22]. Moreover, CCL2 expression was reported to protect cardiac myocytes from cell death in vitro and in vivo [23,48]. This protective effect of CCL2 was attributed to its autocrine effect on cardiomyocytes and its paracrine effects on endothelial cells. The systemic effects of CCL2 in disease mechanisms have also been well studied. While the expression of this chemokine in the heart is our primary motivation, we must understand that any expression at all can lead to a global effect. CCL2 has been implicated in a host of various diseases including inflammatory bowel disease [65], rheumatoid arthritis, diabetes [66], HIV-associated, inflammation-mediated neuronal injury [67,68], ischemia-associated neuronal death [69] and tumor angiogenesis [70]. The hallmark of this chemokine indeed is its excellent ability to recruit proinflammatory cell types to an area of injury or act as a maladaptive protagonist and lead to increased cell damage or death during pathogenic processes. Chemokinebinding protein, Evasin-4, has been shown to selectively inhibit CC-chemokine-mediated bioactivities. In the study done by Braunersreuther et al., intraperitoneal injections of Evasin-4 during MI significantly reduced cardiac injury/inflammation and improved survival in mice [71]. However, as was mentioned earlier, CCL2 has been shown, in fact, to act in prosurvival capacities [48]. In studies by Morimoto et al., cardiac CCL2 overexpression under the control of the a-cardiac myosin heavy chain promoter (MHC/CCL2 transgenic mice) was shown to not only prevent cardiomyocyte death in I/R injury [72] but also prevent cardiac dysfunction and remodeling in the context of MI [73]. The fact that CCL2 was exclusively 525

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

Review

Jarrah & Tarzami

overexpressed in cardiomyocytes may significantly contribute to observed protective effects. It is widely accepted that the loss of cardiac myocytes is a major contributor in the progression of cardiac remodeling and heart failure. As such, the overexpression of CCL2 exclusively in cardiomyocytes may have emphasized protective effects while limiting its ability to recruit proinflammatory cell types, thus limiting a final, detrimental effect. Although these data suggest CCL2/CCR2 is protective during the development and progression of cardiac disease, they also underscore that axis activation in vivo may result in two contradictory effects in the intact heart: death caused by the recruitment of leukocytes and survival from direct cytoprotection or indirect angiogenesis. The net effect of CCL2/ CCR2 signaling in the intact animal lies on the magnitude of this protection relative to CCR2-mediated chemotaxis of potentially damaging inflammatory cells. This interplay clearly delineates why future studies must accurately characterize the autocrine and paracrine signaling events downstream of CCL2/ CCR2 in cardiac myocytes, neutrophils and other heart residence cells. This may provide a means to selectively activate cytoprotective pathways while inhibiting chemotaxis of potentially damaging inflammatory cells, thereby limiting myocardial damage while improving cardiac function. CCL5 (RANTES)

CCL5/RANTES is known to be a very potent chemoattractant for a variety of cell types including T lymphocytes, natural killer cells, monocytes and eosinophils [74,75]. CCL5 is located on human chromosome 17q11.2 and is a more typical ligand in that it binds to multiple receptors, namely CCR1, CCR3, CCR4 and CCR5 [76]. CCL5 is released by many cell types, including astrocytes, endothelial cells, smooth muscle cells, macrophages, activated T cells and platelets [77–80]. CCL5 has been shown to contribute to angiogenesis by stimulating endothelial cell sprouting and tube formation in angiogenesis assays [81]. The incubation of endothelial cells from different organs with CCL5 resulted in a rearrangement of these cells and the formation of pseudo-vessels of human appendix endothelial cells [82]. The duality of CCL5 expression is associated with the ability to induce leukocyte recruitment and activation [83] and may play a role in both chronic inflammation and wound repair [84]. CCL5 has been reported to regulate the inflammatory pathways that promote atherosclerosis; it is highly expressed in human transplant-associated accelerated atherosclerosis [75]. Blocking CCL5-mediated signaling in vivo resulted in a significant reduction of atherosclerosis in animal models tested [75]. Moreover, different genetic polymorphisms of CCL5 are associated with susceptibility to coronary artery disease (CAD) and stent restenosis; these polymorphisms were discovered in the promoter region of CCL5 [85]. CCL5-403A allele polymorphism was associated with the presence and severity of CAD independently of conventional cardiovascular risk factors, eventually leading to clinically significant phenotype alterations. 526

In the context of cardiovascular disease, two fairly recent studies have shown that CCL5 may play a leading role in the death of the injured myocardium. In the murine model of MI used by Montecucco et al., researchers saw that the inhibition of CCL5 using a monoclonal antibody (mAb) resulted in a smaller infarct size 24 h post-MI and increased general survival and cardiac function at the conclusion of the study (21 days) [45]. Braunersreuther et al. observed a similar result in their I/R model; intraperitoneal injection of a CCL5 antagonist leads to an overall decrease in the levels of complimentary chemokines, namely CCL2, and perhaps more importantly oxidative stress and cardiomyocyte death [86]. Thus, the beneficial effect of an anti-CCL5 mAb was associated with reduced chemotaxis of potentially damaging inflammatory cells into the reperfused myocardium, as well as a decrease in the expression of CCL2, oxidative stress and apoptosis [86]. Indeed most of the published data on cardiac disease indicate that CCL5 inhibition may exert cardioprotective effects during early myocardial reperfusion through its anti-inflammatory properties. Interestingly, the prosurvival roles of CCL5 are also reported in brain development; CCL5 promotes the growth and survival of human first-trimester forebrain astrocytes [87]. Moreover, IFN-g, a cytokine that is critical for innate and adaptive immunity, was required for CCL5 effects because blocking IFN-g with an antibody reversed the effects of CCL5. These findings yet again serve as another example of how intertwined the cytokine/chemokine networks can be. Chemokines and their receptors can function directly on some cell types but they can also indirectly affect cell fates through the activation of other cytokines expression, that is, IFN-g. CXCL2 (MIP-2)

CXCL2/MIP-2 was first characterized in 1989 by Cerami et al.; CXCL2 was shown to be a functional murine homologue of human CXCL8, a close relative of CXCL1 and a chemokine that is a primary activator for polymorphonuclear leukocytes [88]. Further studies have shown that monocytes and macrophages are the main sources of CXCL2, which is chemotactic for hematopoietic stem cells in addition to polymorphonuclear leukocytes [89,90]. CXCL2 is located on human chromosome 4q21.1 and associates with its only known receptor, CXCR2 [76]. We note that functional and structural similarities exist between CXCL2 and other a-chemokines such as CXCL1, -5, -6 and -8. CXCL2 belongs to the group of CXCchemokines that have a specific amino acid sequence of glutamic acid-leucine-arginine (abbr. ELR) immediately before the first cysteine of the CXC-motif (ELR+). ELR+ CXCchemokines specifically induce the migration of neutrophils, and the presence of this motif is associated with a proangiogenic function [91]. As we should suspect, CXCL2 has been implicated in a number of systemic diseases. Wu et al. demonstrated the increased expression of CXCL2, as regulated by the differential expression of microRNAs in a swath of gastrointestinal diseases including ulcerative colitis and Crohn’s disease [92], CAD [93] Expert Rev. Clin. Immunol. 11(4), (2015)

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

Chemokines in heart failure

and I/R injury [94]. It has become apparent over the past years that many chemokine receptors display marked promiscuity of ligand binding, which greatly complicates studying their potential roles in a disease. For example, CXCR2 also binds to the cytokine macrophage migration inhibitory factor (MIF), which plays a critical role in inflammatory diseases and atherogenesis. Abrogation of MIF binding, but not CXCL2, the canonical ligand of CXCR2, resulted in a significant reduction of atherosclerosis and reduced monocyte infiltrate in the plaques of animal models tested [93], implicating that atherogenic or inflammatory monocyte recruitment induced by CXCR2 relies not only on CXCL2 binding but also on MIF binding. In I/R injury, post-ischemic inflammatory reaction is a major determinant of the extent of cardiac injury and CXCL2 is considered an important mediator involved in I/R injury through modulating neutrophil recruitment [95]. Neutrophil release of proteolytic enzymes, reactive oxygen species and inflammatory cytokines have been identified as the main culprits in mediating organ damage in early phases of reperfusion [95]. Selectively neutralizing CXC-chemokine bioactivity, for example, CXCL2, using chemokine-binding protein (Evasin-3) during MI, significantly reduced the infarct size by preventing neutrophil recruitment and reactive oxygen species production in myocardial I/R [95]. In contrast however, our laboratory and others have shown that CXCL2 has protective effects in the setting of ischemia/hypoxia injury [23,96]. CXCL2 is suggested as an effective preconditioning therapy against increasingly severe ischemic episodes [96]. CXCL2 overexpression inhibits myocyte apoptosis in vitro, but opposite effects are also observed in vivo; protection seems to have been masked by infiltrating inflammatory cells [23]. The fact that CXCL2 is expressed in cardiac cells and can differentially regulate cardiac cell survival but simultaneously recruit inflammatory cells that may potentially cause cardiac injury post-reperfusion reiterates the point that cardiomyocytespecific overexpression of CXCL2 might be advantageous as it would bypass potential inflammatory roles. CXCL12 (SDF-1)

CXCL12, along with one of its receptors CXCR4, is one of the most studied ligand–receptor pairs [97]. While technically a CXC-chemokine due to an intervening cysteine residue at the N-terminus, typical of CXC’s, the primary structure of CXCL12 appears to be equally related to C-C chemokines [98,99]. CXCL12 is located on human chromosome 10q11.21. Even though CXCL12 is an ELR-chemokine, and is expected to have angiostatic properties, it is well known that CXCL12 is angiogenic [100]. This chemokine is critical to fetal development given that deletion of either the ligand [101] or receptor [102] results in an embryonic lethal mutation resulting from cardiovascular septal and gastrointestinal vascular defects; deficiencies in hematopoiesis and myelopoiesis and deficiencies in neutrophil and B-cell production. This finding coupled with the phenotypic similarity of CXCL12- or CXCR4-deficient mice suggests that this axis comprises a monogamous signaling unit in vivo [103], which is a marked contrast to the typically informahealthcare.com

Review

promiscuous chemokine axes. We should note here however that recent studies have shown that CXCR7, previously believed to be an orphan receptor, is now known to associate with CXCL12 and has thus been dubbed the CXCL12 alternative receptor [104]. Furthermore, CXCR7 has been shown to heterodimerize with CXCR4, suggesting that CXCR7 may act as a modulator of this axis’ signal in various cell types and capacities [105]. CXCL12 is constitutively produced by stromal, dendritic, myocardial and endothelial cells [30,106,107]; the highest concentrations of this chemokine are found in bone marrow, spleen red pulp and lymph node medulla [108]. Various cell types ubiquitously express CXCR4, including endothelial cells, smooth muscle cells, neurons and cardiomyocytes [30,109–111]. Both CXCR4 and CXCL12 are expressed by and function in cardiomyocytes. Increased expression of this axis has been observed in the myocardium of patients with heart failure, suggesting that there may be cardiomodulatory functions [37,111]. Recently published findings support that CXCR4 may impact b-adrenergic processes such as heart failure progression. Data have shown that this chemokine negatively regulates acute activation of b-adrenergic receptor signaling in vitro inhibiting isoproterenolinduced L-type calcium channel activity, negates phospholamban phosphorylation and limits peak calcium transients and myocyte contraction [111]. These data suggest CXCR4 may impact b-adrenergic processes such as heart failure progression. The broad expression pattern of both receptor and ligand make tight control of receptor responsiveness necessary to maintain signaling specificity. CXCL12/CXCR4 plays an important role post-acute MI yielding a potent chemotactic signal for recruiting circulating CXCR4+ progenitor cells to the damaged myocardium. However, the increased CXCL12 signal is transient, lasting approximately 24 h post-MI [37,112]. Even though the CXCL12/CXCR4 axis has been shown to recruit stem cells and promote neovascularization post-MI, it was also reported that the selective inhibition of its receptor, CXCR4, by the antagonist AMD3100, reduced scar formation and enhanced cardiac contractility [49]. Yet Dai et al. recently reported that long-term AMD3100 (CXCR4 specific antagonist) administration leads to increased scar size and adverse ventricular remodeling in murine models of chronic MI, further implicating the beneficial effect of the CXCR4 axis post-MI [113]. Finally, adenoviral overexpression of CXCR4 exacerbated cardiac dysfunction in a rat model of I/R injury through augmentation of TNF-a expression and inflammation [114]. Given the keen interest in using CXCL12 as a therapeutic target in cardiovascular disease [115,116], in order to better understand the protective versus maladaptive effects of CXCL12 and CXCR4 in cardiac injury models, it is essential to determine the direct effects of CXCL12 and its receptor, CXCR4, on myocardial function at the cellular level. As we further define the role of the CXCL12/CXCR4 in the heart, we should note that the axis is also well studied in the context of HIV infection. HIV utilizes CXCR4 as a co-receptor with CCR5 [117,118]. Heart muscle disease is the most important 527

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

Review

Jarrah & Tarzami

cardiovascular manifestation of HIV infection [119,120]. This may take the form of either a dilated cardiomyopathy [121,122] or isolated left or right ventricular dysfunction, is associated with a poor prognosis, and results in symptomatic heart failure in up to 5% of HIV patients [119,123]. Autopsy studies have shown that up to 25% of HIV-infected adults have dilated cardiomyopathy and up to 52% have myocarditis [123]. WHIM syndrome is the first disease identified in humans due to a mutation in a chemokine receptor [124]. The acronym WHIM syndrome was coined to describe an unusual form of congenital neutropenia characterized by warts, hypogammaglobulinemia, immunodeficiency and myelokathexis (the apparent retention of mature neutrophils in the bone marrow) [125]. WHIM syndrome is caused by heterozygous truncating mutations in the cytoplasmic tail domain of the gene encoding CXCR4 [126]. The CXCR4 WHIM mutant fails to internalize upon CXCL12 binding. This constitutive signaling event results in increased retention of neutrophils in the bone marrow and enhanced migration towards CXCL12 [127–129]. Family members of WHIM patients have not undergone systematic cardiac evaluations. There is, however, a 10% incidence of cardiac abnormalities, the most common of which are ventricular septal defects, pulmonary atresia and tetralogy of Fallot [124,130]. The overwhelming majority of aforementioned studies have focused on chemokine expression as a prominent feature of the post-infarction inflammatory response and therefore solely on chemokines’ role in inflammatory leukocyte recruitment [131]. These studies have not addressed the possibility of an autocrine/paracrine effect wherein the chemokine receptors, present on the cardiomyocytes surface, modulate functional responses to stress. During the past decade, a number of studies have elucidated some of the mechanisms by which inflammatory mediators cause heart dysfunction. Their works, regardless of whether cytokines or chemokines were questioned as the mediators of negative inotropy, have suggested paracrine pathways. As we delve deeper, the roles of cytokines and chemokines in scores of clinical conditions will continue to be clarified. These targets could have profound implications in disease management and treatment; which has however been a difficult problem to address in humans due to an apparent lack of genetic abnormalities involving chemokines and their receptors. Stem cells & stem cell-based cytokine therapies

While there is still much unknown, within the past 20 years, there has been a tremendous effort from all walks of the scientific community to characterize and mitigate diseases of the human heart. As a strategy for repairing damaged cardiac tissue following MI, one potentially promising approach involves the use of cell therapy [132–136] to prevent or reverse heart failure (FIGURE 1) [137]. Work began with committed cells such as skeletal myoblasts [138], but recently the field has expanded to explore a wider range of cell types. Most types of stem cells that have been implemented in clinical practice are bone marrowmononuclear cells, BM-MSCs (bone-marrow-derived mesenchymal stem cells), adipose-derived stem cells and endothelial 528

progenitor cells. However, these stem cells yield a very modest improvement in cardiac function [133,139–145]. Endogenous cardiac stem or progenitor cells have also been considered important targets to explore, and three clinical trials (SCIPIO [146], CADUCEUS [147] and ALCADIA [148]) have been conducted to test the therapeutic efficacy of these cells for ischemic heart disease. Even with encouraging initial results, however, controversy still exists regarding the ability of these cells to acquire cardiac cell lineages, promote myocardial regeneration and reconstitute the amount of mass lost after infarction. Interestingly, there are multiple studies in the literature which demonstrate that the effect of adult cell therapy is enacted by the release of paracrine factors that promote repair by mobilizing endogenous progenitors, that is, cytokines, chemokines and growth factors [112,149–152]. One such example is the study by Marba´n et al. which demonstrated that cardiosphere-derived cells are capable of secreting large amounts of angiopoietin-2, bFGF, HGF, IGF-1, VEGF and CXCL12. These findings suggest that upside effects seen in the CADUCEUS trial, which utilized cardiosphere-derived cells, may be due to the secretion of the paracrine factors which in turn stimulated cardiac repair post-MI [152]. The question still remains however: “should we administer cells or cell products to promote cardiac repair and/or mobilize endogenous cardiac precursors?” This has led to a recent clinical trial (ACRX-100) in which researchers attempted to mobilize a patients’ own cardiac stem cells using CXCL12 gene therapy as a homing signal [153]. In this study, researchers used the CXCL12 to draw stem cells to the site of injury and enhance the body’s stem cell-based repair process [154]. This study was different from other cell therapies that typically involve extracting and expanding the number of cells, then delivering them back to the subject. Even though the results of ACRX-100 seemed promising, there are some concerns regarding the results: they had a very small group of participants; and there was no placebo group. Although this approach could be promising for patients with ischemic heart disease, the potential benefits of these findings must be accepted with caution. We should note that current research is now focused on using stem cell technology/transplantation to explore possible tissue regeneration capabilities. A major hurdle in the stem cell therapy is the optimization of such techniques; therapies are not yet able to safely transplant a significant number of cells into the myocardium at the site of injury or infarct. There are many animal models that are currently being used in preclinical experimental models, however, limitations have been identified in all. The main approaches currently being used to deliver these immune mediators, (stem-) cells and/or vectors, in preclinical animal studies of acute MI, include intramyocardial injections, epicardial deposition and intracoronary or transvascular application. Published data comparing intravenous, intracoronary and endocardial delivery of MSCs immediately following infarction in a paracrine model suggests that intracoronary delivery of MSCs was associated with a significantly greater number of engrafted MSCs compared to either Expert Rev. Clin. Immunol. 11(4), (2015)

Chemokines in heart failure

Review

Stem cell renewal

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

3) Cardiac differentiation

Cardiac stem cells

CXCL12

BM-derived stem cells

Cardiac myocytes

Myocardial regeneration Exogenous chemokines/ growth factors and/or stem cells

Damaged myocardium 2) Cardiac stem cells mobilization CXCL12

Acute MI: Endogenous CXCL12 released from heart 1) BM-derived stem cells mobilization

Figure 1. Stem cell-based cytokine therapy: in acute myocardial ischemia (MI), cardiac repair involves stem cell mobilization, retention, and trans-differentiation into functional cardiomyocytes. Upregulation of CXCL12 can act as a homing signal to further assist the recruitment of these stem cells.

percutaneous endocardial delivery or intravenous delivery. Nevertheless, the percentage of retained/engrafted cells was low in all three groups [155,156]. The biggest advantage of using direct injection is that it helps to localize implanted cells into the infarct region and improves graft retention. However, successful chemokine protein deliveries are affected by barriers including the rapid diffusion of chemokines and proteolytic cleavage of chemokines by proteases that are activated in injured tissues [157]. To circumvent this drawback, the epicardium and pericardial sac may offer a less invasive approach for intrapericardial delivery. The pericardium encloses the whole heart, creating a small fluidfilled compartment; this offers an ideal environment for local delivery of (stem-) cells and/or vectors to the heart. However, migration of cells across the visceral pericardium into the myocardium is necessary for the success of this approach. Moreover, there is little known about the efficiency and efficacy of cells delivered via this approach. The molecular mechanisms for stem cell mobilization have not been identified. It has been assumed that stem cells migrate similarly to leukocytes, even though, there has been little evidence supporting it. Future studies will be needed to optimize a proper transmigration and informahealthcare.com

subsequent cross-talk between the engrafted cells and the host myocardium. For patients presenting an acute MI, the intracoronary delivery approach will be the less invasive approach and can be accomplished easily following catheter intervention to reestablish coronary flow. The results of preclinical animal studies for intrapericardial delivery have been very promising; in particular, using cytokine/chemokine seeded biomaterials for intrapericardial delivery. This approach allows for a time-controlled release of these proteins using biodegradable hydrogels which presents a potential advantage [158]. Cytokine-based immunotherapy: anti-cytokine therapies

Cytokine-based immunotherapies are widely used in cancer treatment, where the immune system is harnessed to eradicate cancer [159,160]. Animal studies have shown that cytokines have broad antitumor activity; a cytokine-coordinated response to a target antigen has been well translated into a number of cytokine-based immunotherapy approaches for cancer [161,162]. In spite of this, there is overwhelming evidence to support that the immune mediators (inflammatory cytokines and 529

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

Review

Jarrah & Tarzami

chemokines) can contribute to cardiac dysfunction and the progression of heart failure by promoting cardiac hypertrophy, cell death and/or fibrosis. As such, cytokine-based immunotherapies haven not been yet successful in the treatment of heart failure. This may be partially due to the fact that chronic heart failure is a multifactorial disease; in addition to the immune system, there are implications in a variety of body mechanisms including the neurohormonal system, which are known to activate in response to the heart failure state. Moreover, activation of the neurohormonal systems in response to cardiac injury can have deleterious effects on the heart, which ultimately causes the disease to progress [163,164]. Since the cytokine network is compromised of both inflammatory and anti-inflammatory immune mediators, any inflammation associated with aforementioned diseases might be the result of an imbalance of the two opposing arms of the cytokine network: an increase in inflammatory cytokines and/or a decrease of anti-inflammatory cytokines. Therefore, to create an equilibrium, immunotherapies for heart failure have to target inflammatory mechanisms in a way that decreases the relative ratio of inflammatory cytokines to anti-inflammatory cytokines. Based on this premise, the ACCLAIM trial (Celacade immunotherapy) was designed using a novel non-drug treatment strategy to specifically downregulate proinflammatory cytokines and increase anti-inflammatory cytokines in heart failure patients [165]. The trial was reported safe and well tolerated and seemed to show promise in reducing risk of mortality or morbidity. This trial is a marked contrast from the majority of current efforts, given that inflammatory cytokines are believed to negatively influence contractility and remodeling processes during progression of chronic heart failure. To date, most researchers are more interested in anti-cytokine therapy to improve myocardial performance and which is reflected in the design of past clinical trials (TABLE 1). As we previously mentioned, increased circulating levels of proinflammatory cytokines, that is, TNF-a, IL-1 and IL-6, have been reported within the failing myocardium [16,18,19,33,37,166,167]; among these factors, TNF-a has received the most attention. TNF-a is elevated in the serum of patients with heart failure, and the magnitude of the increase is directly correlated with the severity of disease [16]. Indeed, overproduction of TNF-a by cardiomyocytes is sufficient enough to cause pathological changes in the myocardium consistent with heart failure, including ventricular remodeling, interstitial fibrosis and cardiomyocyte apoptosis [8]. Thus, it was postulated that inhibition of TNF-a might favorably modify the progression of heart failure; however, the ATTACH (Anti-TNF-a Therapy Against Congestive Heart failure) [168] trial failed to show any improvements in patients with moderate-to-severe heart failure. Results of this study were followed up by unfavorable outcomes of other antiTNF-a clinical trials such as the Randomized Etanercept North American Strategy to Study Antagonism of Cytokine (RENASSANCE), the Research into Etanercept: Cytokine Antagonism in Ventricular function (RECOVER) and RENEWAL (combine analysis on RENASSANCE + RECOVER) [11,169–172]. One 530

interpretation of the failure of these anti-cytokine approaches in comparison to other clinical trials [24,173–176] is that the disease is multifactorial in nature; targeting a single component of the inflammatory cascade without regard to the cellular origin or the mechanism underlying the chronic expression is not a sufficient approach. Conclusion

The focus here though has been to describe the parallel process in the intact heart in which the activity of these chemokines/ cytokines can be both protective and detrimental. Summarized are the rather elusive roles of these proteins and how characterizing these receptor–ligand axes are of critical importance if we are to translate their noted success in cancer treatment and produce efficient cardiac therapies. As of yet, our characterizations cannot do much beyond describe these cytokines/chemokines as both protective and detrimental. Aside from the potential in stem cells to promote cardiac repair, current therapies have failed to address autocrine/paracrine effects in this process. Although both views (stem cell-based cytokine therapy and anti-cytokine therapy) have merit in using cytokines/ chemokines as new targets for therapy in myocardial failure, the results of these past clinical trials suggest that we simply have not targeted these mediators with the proper modes when considering the context of heart failure. The question remains unanswered: “Are the previous targets, namely anti-cytokine therapies, still relevant or should we look for novel targets?” We believe that increased attention should be given to how proinflammatory mediators are modulated and what is the underlying cell network that serves both as an important source and effector target for cytokines in order to develop more specific immunomodulating agents to combat chronic heart failure. Furthermore, stem cell-based cytokine therapy may play a pivotal role in cardiac repair, and regardless even of its shortcomings, is a promising approach for patients with acute MI. Drawbacks stem from the fact that stem cell-based therapies are at an early stage and much needs to be done to optimize stem cell mobilization, retention and engraftment into the failing heart. With further research and tuning, however, this approach may hold promise. Expert commentary

The jury is still out on whether or not the future of non-invasive, chemokine-dependent therapies is a rich one; further development and refinement of current strategies may one day lead to impressive and powerful techniques to combat cardiac disease. However, the complexity of these targets leads us to believe that therapies will need to be highly specific, thus posing the question of whether or not pursing such non-general treatments is appropriate. The coming years will be a pivotal point in the progression or expulsion of these treatments. Five-year view

Findings from numerous studies support the feasibility of cardiac repair using stem cell mobilization to the infarcted heart Expert Rev. Clin. Immunol. 11(4), (2015)

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

Chemokines in heart failure

with factors such as cytokines/chemokines; however, these factors have additional direct effects on the recruitment of inflammatory cells into the injured heart, and it is difficult to determine the beneficial contribution of chemokines in stem cell mobilization from those associated with inflammatorymediated injuries. Since many chemokines subfamilies are expressed on cardiac cells, it is hoped that their respective autocrine/paracrine signals in the heart, and more importantly the injured heart, will be characterized in the near future.

Review

This will allow for the development of more adept therapeutic remedies. Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or material discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Key issues .

The present review provides a critical overview of the clinical status of immunotherapy in patients with a failing heart.

.

The duality of chemokines has been at times confounding, yet the ability of the same axis to play opposite roles suggests that these signals operate differently depending on the injury or disease environment.

.

A related strategy for cardiac repair involves stem cell mobilization with factors such as cytokines/chemokines (stem cell-based cytokine therapies).

.

There has been an interest in anti-cytokine therapy to improve myocardial performance; however, past clinical trials have failed to show any clinical benefits.

8.

Bryant D, Becker L, Richardson J, et al. Cardiac failure in transgenic mice with myocardial expression of tumor necrosis factor-alpha. Circulation 1998;97:1375-81

Go AS, Mozaffarian D, Roger VL, et al. Heart disease and stroke statistics – 2014 update: a report from the American Heart Association. Circulation 2014;129: e28-e292

9.

Sivasubramanian N, Coker ML, Kurrelmeyer KM, et al. Left ventricular remodeling in transgenic mice with cardiac restricted overexpression of tumor necrosis factor. Circulation 2001;104:826-31

2.

Kapadia S, Torre-Amione G, Yokoyama T, et al. Soluble TNF binding proteins modulate the negative inotropic properties of TNF-alpha in vitro. Am J Physiol 1995;268:H517-25

10.

3.

van der Poll T, van Deventer SJ. Cytokines and anticytokines in the pathogenesis of sepsis. Infect Dis Clin North Am 1999;13: 413-26; ix

11.

4.

Gulick T, Chung MK, Pieper SJ, et al. Interleukin 1 and tumor necrosis factor inhibit cardiac myocyte beta-adrenergic responsiveness. Proc Natl Acad Sci USA 1989;86:6753-7

References

15.

Berthonneche C, Sulpice T, Boucher F, et al. New insights into the pathological role of TNF-alpha in early cardiac dysfunction and subsequent heart failure after infarction in rats. Am J Physiol Heart Circ Physiol 2004;287:H340-50

16.

Levine B, Kalman J, Mayer L, et al. Elevated circulating levels of tumor necrosis factor in severe chronic heart failure. N Engl J Med 1990;323:236-41

17.

Ferrari R, Bachetti T, Confortini R, et al. Tumor necrosis factor soluble receptors in patients with various degrees of congestive heart failure. Circulation 1995;92:1479-86

Kadokami T, McTiernan CF, Kubota T, et al. Effects of soluble TNF receptor treatment on lipopolysaccharide-induced myocardial cytokine expression. Am J Physiol Heart Circ Physiol 2001;280: H2281-91

18.

Testa M, Yeh M, Lee P, et al. Circulating levels of cytokines and their endogenous modulators in patients with mild to severe congestive heart failure due to coronary artery disease or hypertension. J Am Coll Cardiol 1996;28:964-71

12.

Yokoyama T, Nakano M, Bednarczyk JL, et al. Tumor necrosis factor-alpha provokes a hypertrophic growth response in adult cardiac myocytes. Circulation 1997;95: 1247-52

.

Data from this study suggests that in patients with congestive heart failure, circulating levels of cytokines increased with the severity of symptoms.

19.

13.

Kadokami T, McTiernan CF, Kubota T, et al. Sex-related survival differences in murine cardiomyopathy are associated with differences in TNF-receptor expression. J Clin Invest 2000;106:589-97

Aukrust P, Ueland T, Muller F, et al. Elevated circulating levels of C-C chemokines in patients with congestive heart failure. Circulation 1998;97:1136-43

20.

Kukielka GL, Youker KA, Michael LH, et al. Role of early reperfusion in the induction of adhesion molecules and cytokines in previously ischemic myocardium. Mol Cell Biochem 1995;147: 5-12

21.

Martin AP, Coronel EC, Sano G, et al. A novel model for lymphocytic infiltration

Papers of special note have been highlighted as: . of interest .. of considerable interest 1.

5.

6.

7.

Finkel MS, Hoffman RA, Shen L, et al. Interleukin-6 (IL-6) as a mediator of stunned myocardium. Am J Cardiol 1993;71:1231-2 McGowan FX Jr, Takeuchi K, del Nido PJ, et al. Myocardial effects of interleukin-2. Transplant Proc 1994;26:209-10 Kubota T, McTiernan CF, Frye CS, et al. Dilated cardiomyopathy in transgenic mice with cardiac-specific overexpression of tumor necrosis factor-alpha. Circ Res 1997;81:627-35

informahealthcare.com

14.

Chung MK, Gulick TS, Rotondo RE, et al. Mechanism of cytokine inhibition of betaadrenergic agonist stimulation of cyclic AMP in rat cardiac myocytes. Impairment of signal transduction. Circ Res 1990;67:753-63

Kubota T, Miyagishima M, Alvarez RJ, et al. Expression of proinflammatory cytokines in the failing human heart: comparison of recent-onset and end-stage congestive heart failure. J Heart Lung Transplant 2000;19:819-24

531

Review

Jarrah & Tarzami

of the thyroid gland generated by transgenic expression of the CC chemokine CCL21. J Immunol 2004;173:4791-8 22.

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

23.

.

24.

Hong KH, Ryu J, Han KH. Monocyte chemoattractant protein-1-induced angiogenesis is mediated by vascular endothelial growth factor-A. Blood 2005;105:1405-7

Smolen J, Landewe RB, Mease P, et al. Efficacy and safety of certolizumab pegol plus methotrexate in active rheumatoid arthritis: the RAPID 2 study. A randomised controlled trial. Ann Rheum Dis 2009;68: 797-804

25.

Thompson MD, Burnham WM, Cole DE. The G protein-coupled receptors: pharmacogenetics and disease. Crit Rev Clin Lab Sci 2005;42:311-92

26.

Tang CM, Insel PA. Genetic variation in G-protein-coupled receptors – consequences for G-protein-coupled receptors as drug targets. Expert Opin Ther Targets 2005;9: 1247-65

27.

Hopkins AL, Groom CR. The druggable genome. Nat Rev Drug Discov 2002;1: 727-30

28.

Murdoch C, Finn A. Chemokine receptors and their role in inflammation and infectious diseases. Blood 2000;95:3032-43

29.

Brouty-Boye D, Pottin-Clemenceau C, Doucet C, et al. Chemokines and CD40 expression in human fibroblasts. Eur J Immunol 2000;30:914-19

30.

31.

32.

33.

34.

Tarzami ST, Cheng R, Miao W, et al. Chemokine expression in myocardial ischemia: MIP-2 dependent MCP-1 expression protects cardiomyocytes from cell death. J Mol Cell Cardiol 2002;34:209-21 Data from this study demonstrated for the first time that chemokines and their respective receptors are expressed in the cardiomyocytes and they have distinct physiological functions.

Gupta SK, Lysko PG, Pillarisetti K, et al. Chemokine receptors in human endothelial cells. Functional expression of CXCR4 and its transcriptional regulation by inflammatory cytokines. J Biol Chem 1998;273:4282-7 Schecter AD, Calderon TM, Berman AB, et al. Human vascular smooth muscle cells possess functional CCR5. J Biol Chem 2000;275:5466-71 Hohensinner PJ, Kaun C, Rychli K, et al. Monocyte chemoattractant protein (MCP-1) is expressed in human cardiac cells and is differentially regulated by inflammatory

532

42.

Damas JK, Gullestad L, Ueland T, et al. CXC-chemokines, a new group of cytokines in congestive heart failure – possible role of platelets and monocytes. Cardiovasc Res 2000;45:428-36

Kolattukudy PE, Quach T, Bergese S, et al. Myocarditis induced by targeted expression of the MCP-1 gene in murine cardiac muscle. Am J Pathol 1998;152:101-11

43.

Shioi T, Matsumori A, Kihara Y, et al. Increased expression of interleukin-1 beta and monocyte chemotactic and activating factor/monocyte chemoattractant protein-1 in the hypertrophied and failing heart with pressure overload. Circ Res 1997;81:664-71

Hayashidani S, Tsutsui H, Shiomi T, et al. Anti-monocyte chemoattractant protein-1 gene therapy attenuates left ventricular remodeling and failure after experimental myocardial infarction. Circulation 2003;108:2134-40

44.

Devaux B, Scholz D, Hirche A, et al. Upregulation of cell adhesion molecules and the presence of low grade inflammation in human chronic heart failure. Eur Heart J 1997;18:470-9

45.

Behr TM, Wang X, Aiyar N, et al. Monocyte chemoattractant protein-1 is upregulated in rats with volume-overload congestive heart failure. Circulation 2000;102:1315-22

Montecucco F, Braunersreuther V, Lenglet S, et al. CC chemokine CCL5 plays a central role impacting infarct size and post-infarction heart failure in mice. Eur Heart J 2012;33:1964-74

46.

Dama˚s JK, Eiken HG, Oie E, et al. Myocardial expression of CC- and CXC-chemokines and their receptors in human end-stage heart failure. Cardiovasc Res 2000;47:778-87

Kaikita K, Hayasaki T, Okuma T, et al. Targeted deletion of CC chemokine receptor 2 attenuates left ventricular remodeling after experimental myocardial infarction. Am J Pathol 2004;165:439-47

47.

Suffee N, Hlawaty H, Meddahi-Pelle A, et al. RANTES/CCL5-induced pro-angiogenic effects depend on CCR1, CCR5 and glycosaminoglycans. Angiogenesis 2012;15:727-44

48.

Tarzami ST, Calderon TM, Deguzman A, et al. MCP-1/CCL2 protects cardiac myocytes from hypoxia-induced apoptosis by a G(alphai)-independent pathway. Biochem Biophys Res Commun 2005;335: 1008-16

49.

Liehn EA, Tuchscheerer N, Kanzler I, et al. Double-edged role of the CXCL12/ CXCR4 axis in experimental myocardial infarction. J Am Coll Cardiol 2011;58: 2415-23

50.

Van Coillie E, Van Damme J, Opdenakker G. The MCP/eotaxin subfamily of CC chemokines. Cytokine Growth Factor Rev 1999;10:61-86

51.

Craig MJ, Loberg RD. CCL2 (Monocyte Chemoattractant Protein-1) in cancer bone metastases. Cancer Metastasis Rev 2006;25: 611-19

52.

Deshmane SL, Kremlev S, Amini S, et al. Monocyte chemoattractant protein-1 (MCP1): an overview. J Interferon Cytokine Res 2009;29:313-26

53.

Rollins BJ. Chemokines. Blood 1997;90: 909-28

.

This article contains a comprehensive review on chemokines.

mediators and hypoxia. FEBS Lett 2006;580:3532-8

35.

36.

37.

.

38.

Seino Y, Ikeda U, Sekiguchi H, et al. Expression of leukocyte chemotactic cytokines in myocardial tissue. Cytokine 1995;7:301-4

Results from this study suggests that the expression of chemokines and their respective receptors are dysregulated in human end-stage heart failure suggesting their potential involvement in progression of chronic heart failure. Tarzami ST, Miao W, Mani K, 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.

Talvani A, Rocha MO, Barcelos LS, et al. Elevated concentrations of CCL2 and tumor necrosis factor-alpha in chagasic cardiomyopathy. Clin Infect Dis 2004;38: 943-50

40.

Fuse K, Kodama M, Hanawa H, et al. Enhanced expression and production of monocyte chemoattractant protein-1 in myocarditis. Clin Exp Immunol 2001;124: 346-52

41.

Damas JK, Gullestad L, Aass H, et al. Enhanced gene expression of chemokines and their corresponding receptors in mononuclear blood cells in chronic heart failure – modulatory effect of intravenous immunoglobulin. J Am Coll Cardiol 2001;38:187-93

Expert Rev. Clin. Immunol. 11(4), (2015)

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

Chemokines in heart failure

54.

Rollins BJ, Morrison ED, Stiles CD. Cloning and expression of JE, a gene inducible by platelet-derived growth factor and whose product has cytokine-like properties. Proc Natl Acad Sci USA 1988;85:3738-42

55.

Kim MS, Day CJ, Morrison NA. MCP-1 is induced by receptor activator of nuclear factor-{kappa}B ligand, promotes human osteoclast fusion, and rescues granulocyte macrophage colony-stimulating factor suppression of osteoclast formation. J Biol Chem 2005;280:16163-9

56.

57.

Banisadr G, Gosselin RD, Mechighel P, et al. Highly regionalized neuronal expression of monocyte chemoattractant protein-1 (MCP-1/CCL2) in rat brain: evidence for its colocalization with neurotransmitters and neuropeptides. J Comp Neurol 2005;489:275-92 Pype JL, Dupont LJ, Menten P, et al. Expression of monocyte chemotactic protein (MCP)-1, MCP-2, and MCP-3 by human airway smooth-muscle cells. Modulation by corticosteroids and T-helper 2 cytokines. Am J Respir Cell Mol Biol 1999;21:528-36

58.

Xia M, Sui Z. Recent developments in CCR2 antagonists. Expert Opin Ther Pat 2009;19:295-303

59.

Matsumori A, Furukawa Y, Hashimoto T, et al. Plasma levels of the monocyte chemotactic and activating factor/monocyte chemoattractant protein-1 are elevated in patients with acute myocardial infarction. J Mol Cell Cardiol 1997;29:419-23

60.

Satoh M, Akatsu T, Ishkawa Y, et al. A novel activator of C-C chemokine, FROUNT, is expressed with C-C chemokine receptor 2 and its ligand in failing human heart. J Card Fail 2007;13: 114-19

61.

Gerszten RE, Mach F, Sauty A, et al. Chemokines, leukocytes, and atherosclerosis. J Lab Clin Med 2000;136:87-92

62.

Gu L, Okada Y, Clinton SK, et al. Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice. Mol Cell 1998;2:275-81

63.

64.

65.

Spoettl T, Hausmann M, Herlyn M, et al. Monocyte chemoattractant protein-1 (MCP1) inhibits the intestinal-like differentiation of monocytes. Clin Exp Immunol 2006;145:190-9

66.

Marisa C, Lucci I, Di Giulio C, et al. MCP-1 and MIP-2 expression and production in BB diabetic rat: effect of chronic hypoxia. Mol Cell Biochem 2005;276:105-11

67.

Ragin AB, Wu Y, Storey P, et al. Monocyte chemoattractant protein-1 correlates with subcortical brain injury in HIV infection. Neurology 2006;66:1255-7

68.

Flora G, Pu H, Lee YW, et al. Proinflammatory synergism of ethanol and HIV-1 Tat protein in brain tissue. Exp Neurol 2005;191:2-12

69.

Sakurai-Yamashita Y, Shigematsu K, Yamashita K, et al. Expression of MCP-1 in the hippocampus of SHRSP with ischemia-related delayed neuronal death. Cell Mol Neurobiol 2006;26:823-31

70.

71.

72.

73.

Wang J, Ou ZL, Hou YF, et al. Enhanced expression of Duffy antigen receptor for chemokines by breast cancer cells attenuates growth and metastasis potential. Oncogene 2006;25:7201-11 Braunersreuther V, Montecucco F, Pelli G, et al. Treatment with the CC chemokine-binding protein Evasin-4 improves post-infarction myocardial injury and survival in mice. Thromb Haemost 2013;110:807-25 Morimoto H, Hirose M, Takahashi M, et al. MCP-1 induces cardioprotection against ischaemia/reperfusion injury: role of reactive oxygen species. Cardiovasc Res 2008;78:554-62 Morimoto H, Takahashi M, Izawa A, et al. Cardiac overexpression of monocyte chemoattractant protein-1 in transgenic mice prevents cardiac dysfunction and remodeling after myocardial infarction. Circ Res 2006;99:891-9

Review

77.

Cota M, Kleinschmidt A, Ceccherini-Silberstein F, et al. Upregulated expression of interleukin-8, RANTES and chemokine receptors in human astrocytic cells infected with HIV-1. J Neurovirol 2000;6:75-83

78.

Shukaliak JA, Dorovini-Zis K. Expression of the beta-chemokines RANTES and MIP-1 beta by human brain microvessel endothelial cells in primary culture. J Neuropathol Exp Neurol 2000;59:339-52

79.

John M, Hirst SJ, Jose PJ, et al. Human airway smooth muscle cells express and release RANTES in response to T helper 1 cytokines: regulation by T helper 2 cytokines and corticosteroids. J Immunol 1997;158:1841-7

80.

von Luettichau I, Nelson PJ, Pattison JM, et al. RANTES chemokine expression in diseased and normal human tissues. Cytokine 1996;8:89-98

81.

Bernardini G, Ribatti D, Spinetti G, et al. Analysis of the role of chemokines in angiogenesis. J Immunol Methods 2003;273:83-101

82.

Crola Da Silva C, Lamerant-Fayel N, Paprocka M, et al. Selective human endothelial cell activation by chemokines as a guide to cell homing. Immunology 2009;126:394-404

83.

Aizenstein HJ, Nebes RD, Meltzer CC, et al. The relation of White Matter Hyperintensities to implicit learning in healthy older adults. Int J Geriatr Psychiatry 2002;17:664-9

84.

Shahrara S, Amin MA, Woods JM, et al. Chemokine receptor expression and in vivo signaling pathways in the joints of rats with adjuvant-induced arthritis. Arthritis Rheum 2003;48:3568-83

85.

Tavakkoly-Bazzaz J, Amiri P, Tajmir-Riahi M, et al. RANTES gene mRNA expression and its -403 G/ A promoter polymorphism in coronary artery disease. Gene 2011;487:103-6

74.

86.

Lehmann MH, Kuhnert H, Muller S, et al. Monocyte chemoattractant protein 1 (MCP1) gene expression in dilated cardiomyopathy. Cytokine 1998;10:739-46

von Hundelshausen P, Weber KS, Huo Y, et al. RANTES deposition by platelets triggers monocyte arrest on inflamed and atherosclerotic endothelium. Circulation 2001;103:1772-7

Braunersreuther V, Pellieux C, Pelli G, et al. Chemokine CCL5/RANTES inhibition reduces myocardial reperfusion injury in atherosclerotic mice. J Mol Cell Cardiol 2010;48:789-98

75.

87.

Ono K, Matsumori A, Furukawa Y, et al. Prevention of myocardial reperfusion injury in rats by an antibody against monocyte chemotactic and activating factor/monocyte chemoattractant protein-1. Lab Invest 1999;79:195-203

Pattison JM, Nelson PJ, Huie P, et al. RANTES chemokine expression in transplant-associated accelerated atherosclerosis. J Heart Lung Transplant 1996;15:1194-9

Bakhiet M, Tjernlund A, Mousa A, et al. RANTES promotes growth and survival of human first-trimester forebrain astrocytes. Nat Cell Biol 2001;3:150-7

88.

76.

Bacon K, Baggiolini M, Broxmeyer H, et al. Chemokine/chemokine receptor nomenclature. J Interferon Cytokine Res 2002;22:1067-8

Wolpe SD, Sherry B, Juers D, et al. Identification and characterization of macrophage inflammatory protein 2. Proc Natl Acad Sci USA 1989;86:612-16

informahealthcare.com

533

Review 89.

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

90.

91.

92.

93.

94.

95.

96.

Jarrah & Tarzami

Iida N, Grotendorst GR. Cloning and sequencing of a new gro transcript from activated human monocytes: expression in leukocytes and wound tissue. Mol Cell Biol 1990;10:5596-9

101.

Pelus LM, Fukuda S. Peripheral blood stem cell mobilization: the CXCR2 ligand GRObeta rapidly mobilizes hematopoietic stem cells with enhanced engraftment properties. Exp Hematol 2006;34:1010-20

102.

Moore BB, Keane MP, Addison CL, et al. CXC chemokine modulation of angiogenesis: the importance of balance between angiogenic and angiostatic members of the family. J Investig Med 1998;46:113-20 Wu F, Zikusoka M, Trindade A, et al. MicroRNAs are differentially expressed in ulcerative colitis and alter expression of macrophage inflammatory peptide-2 alpha. Gastroenterology 2008;135:1624-35.e24 Bernhagen J, Krohn R, Lue H, et al. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med 2007;13:587-96

Zhu HL, Wei X, Qu SL, et al. Ischemic postconditioning protects cardiomyocytes against ischemia/reperfusion injury by inducing MIP2. Exp Mol Med 2011;43: 437-45

Zou YR, Kottmann AH, Kuroda M, et al. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature 1998;393:595-9

103.

Ma Q, Jones D, Borghesani PR, et al. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice. Proc Natl Acad Sci USA 1998;95:9448-53

104.

Balabanian K, Lagane B, Infantino S, et al. The chemokine SDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes. J Biol Chem 2005;280:35760-6

105.

Levoye A, Balabanian K, Baleux F, et al. CXCR7 heterodimerizes with CXCR4 and regulates CXCL12-mediated G protein signaling. Blood 2009;113:6085-93

106.

Deichmann M, Kronenwett R, Haas R. Expression of the human immunodeficiency virus type-1 coreceptors CXCR-4 (fusin, LESTR) and CKR-5 in CD34+ hematopoietic progenitor cells. Blood 1997;89:3522-8

Farivar AS, Krishnadasan B, Naidu BV, et al. Alpha chemokines regulate direct lung ischemia-reperfusion injury. J Heart Lung Transplant 2004;23:585-91 Montecucco F, Lenglet S, Braunersreuther V, et al. Single administration of the CXC chemokine-binding protein Evasin-3 during ischemia prevents myocardial reperfusion injury in mice. Arterioscler Thromb Vasc Biol 2010;30:1371-7

Nagasawa T, Hirota S, Tachibana K, et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature 1996;382:635-8

107.

Pablos JL, Amara A, Bouloc A, et al. Stromal-cell derived factor is expressed by dendritic cells and endothelium in human skin. Am J Pathol 1999;155:1577-86

108.

Heesen M, Berman MA, Benson JD, et al. Cloning of the mouse fusin gene, homologue to a human HIV-1 co-factor. J Immunol 1996;157:5455-60

109.

Banisadr G, Skrzydelski D, Kitabgi P, et al. Highly regionalized distribution of stromal cell-derived factor-1/CXCL12 in adult rat brain: constitutive expression in cholinergic, dopaminergic and vasopressinergic neurons. Eur J Neurosci 2003;18:1593-606

97.

Murdoch C. CXCR4: chemokine receptor extraordinaire. Immunol Rev 2000;177: 175-84

110.

98.

Sallusto F, Lanzavecchia A, Mackay CR. Chemokines and chemokine receptors in T-cell priming and Th1/Th2-mediated responses. Immunol Today 1998;19:568-74

Banisadr G, Rostene W, Kitabgi P, et al. Chemokines and brain functions. Curr Drug Targets Inflamm Allergy 2005;4: 387-99

111.

99.

Bleul CC, Farzan M, Choe H, et al. The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/fusin and blocks HIV-1 entry. Nature 1996;382:829-33

Pyo RT, Sui J, Dhume A, et al. CXCR4 modulates contractility in adult cardiac myocytes. J Mol Cell Cardiol 2006;41:834-44

112.

100.

Salcedo R, Wasserman K, Young HA, et al. Vascular endothelial growth factor and basic fibroblast growth factor induce expression of CXCR4 on human endothelial cells: in vivo neovascularization induced by stromal-derived factor-1alpha. Am J Pathol 1999;154:1125-35

Zhang M, Mal N, Kiedrowski M, et al. SDF-1 expression by mesenchymal stem cells results in trophic support of cardiac myocytes after myocardial infarction. FASEB J 2007;21:3197-207

534

113.

Dai S, Yuan F, Mu J, et al. Chronic AMD3100 antagonism of SDF-1alpha-CXCR4 exacerbates cardiac

dysfunction and remodeling after myocardial infarction. J Mol Cell Cardiol 2010;49:587-97 114.

Chen J, Chemaly E, Liang L, et al. Effects of CXCR4 gene transfer on cardiac function after ischemia-reperfusion injury. Am J Pathol 2010;176:1705-15

115.

Moore MA, Hattori K, Heissig B, et al. Mobilization of endothelial and hematopoietic stem and progenitor cells by adenovector-mediated elevation of serum levels of SDF-1, VEGF, and angiopoietin-1. Ann N Y Acad Sci 2001;938:36-45; discussion 45-7

.

Findings from this study demonstrated that bone marrow-derived stem and/or progenitor cells can be mobilized in response to elevated levels of cytokines.

116.

Hiasa K, Ishibashi M, Ohtani K, et al. Gene transfer of stromal cell-derived factor1alpha enhances ischemic vasculogenesis and angiogenesis via vascular endothelial growth factor/endothelial nitric oxide synthase-related pathway: next-generation chemokine therapy for therapeutic neovascularization. Circulation 2004;109: 2454-61

117.

Feng Y, Broder CC, Kennedy PE, et al. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science 1996;272:872-7

118.

Alkhatib G, Combadiere C, Broder CC, et al. CC CKR5: a RANTES, MIP-1alpha, MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1. Science 1996;272:1955-8

119.

Currie PF, Goldman JH, Caforio AL, et al. Cardiac autoimmunity in HIV related heart muscle disease. Heart 1998;79:599-604

120.

Barbaro G, Di Lorenzo G, Grisorio B, et al. Cardiac involvement in the acquired immunodeficiency syndrome: a multicenter clinical-pathological study. Gruppo Italiano per lo Studio Cardiologico dei pazienti affetti da AIDS Investigators. AIDS Res Hum Retroviruses 1998;14:1071-7

121.

Herskowitz A, Willoughby SB, Baughman KL, et al. Cardiomyopathy associated with antiretroviral therapy in patients with HIV infection: a report of six cases. Ann Intern Med 1992;116:311-13

122.

Barbaro G, Di Lorenzo G, Grisorio B, et al. Incidence of dilated cardiomyopathy and detection of HIV in myocardial cells of HIV-positive patients. Gruppo Italiano per lo Studio Cardiologico dei Pazienti Affetti da AIDS. N Engl J Med 1998;339:1093-9

Expert Rev. Clin. Immunol. 11(4), (2015)

Chemokines in heart failure

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

123.

Moroni M, Antinori S. HIV and direct damage of organs: disease spectrum before and during the highly active antiretroviral therapy era. AIDS 2003;17(Suppl 1):S51-64

124.

Gulino AV. WHIM syndrome: a genetic disorder of leukocyte trafficking. Curr Opin Allergy Clin Immunol 2003;3:443-50

125.

acute myocardial infarction (TOPCAREAMI): mechanistic insights from serial contrast-enhanced magnetic resonance imaging. Circulation 2003;108:2212-18 137.

Zuelzer WW. “Myelokathexis” – a new form of chronic granulocytopenia. Report of a case. N Engl J Med 1964;270:699-704

Laflamme MA, Zbinden S, Epstein SE, et al. Cell-based therapy for myocardial ischemia and infarction: pathophysiological mechanisms. Annu Rev Pathol 2007;2: 307-39

138.

126.

Hernandez PA, Gorlin RJ, Lukens JN, et al. Mutations in the chemokine receptor gene CXCR4 are associated with WHIM syndrome, a combined immunodeficiency disease. Nat Genet 2003;34:70-4

Jain M, DerSimonian H, Brenner DA, et al. Cell therapy attenuates deleterious ventricular remodeling and improves cardiac performance after myocardial infarction. Circulation 2001;103:1920-7

139.

127.

Taniuchi S, Yamamoto A, Fujiwara T, et al. Dizygotic twin sisters with myelokathexis: mechanism of its neutropenia. Am J Hematol 1999;62:106-11

Orlic D, Kajstura J, Chimenti S, et al. Bone marrow cells regenerate infarcted myocardium. Nature 2001;410:701-5

128.

129.

130.

131.

132.

133.

134.

Balabanian K, Harriague J, Decrion C, et al. CXCR4-tropic HIV-1 envelope glycoprotein functions as a viral chemokine in unstimulated primary CD4+ T lymphocytes. J Immunol 2004;173:7150-60 Bachelerie F. CXCL12/CXCR4-axis dysfunctions: markers of the rare immunodeficiency disorder WHIM syndrome. Dis Markers 2010;29:189-98 Badolato R, Dotta L, Tassone L, et al. Tetralogy of Fallot is an uncommon manifestation of warts, hypogammaglobulinemia, infections, and myelokathexis syndrome. J Pediatr 2012;161:763-5 Frangogiannis NG, Entman ML. Targeting the chemokines in myocardial inflammation. Circulation 2004;110:1341-2 Limbourg FP, Ringes-Lichtenberg S, Schaefer A, et al. Haematopoietic stem cells improve cardiac function after infarction without permanent cardiac engraftment. Eur J Heart Fail 2005;7:722-9 Beltrami AP, Barlucchi L, Torella D, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003;114:763-76 Kocher AA, Schuster MD, Szabolcs MJ, et al. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med 2001;7:430-6

135.

Hughes S. Cardiac stem cells. J Pathol 2002;197:468-78

136.

Britten MB, Abolmaali ND, Assmus B, et al. Infarct remodeling after intracoronary progenitor cell treatment in patients with

informahealthcare.com

140.

Murry CE, Soonpaa MH, Reinecke H, et al. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 2004;428:664-8

141.

Toma C, Pittenger MF, Cahill KS, et al. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation 2002;105:93-8

142.

143.

144.

Oh H, Bradfute SB, Gallardo TD, et al. Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc Natl Acad Sci USA 2003;100:12313-18 Mummery C, Ward-van Oostwaard D, et al. Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation 2003;107:2733-40 Mirotsou M, Jayawardena TM, Schmeckpeper J, et al. Paracrine mechanisms of stem cell reparative and regenerative actions in the heart. J Mol Cell Cardiol 2011;50:280-9

145.

Wang WE, Chen X, Houser SR, et al. Potential of cardiac stem/progenitor cells and induced pluripotent stem cells for cardiac repair in ischaemic heart disease. Clin Sci (Lond) 2013;125:319-27

146.

Bolli R, Chugh AR, D’Amario D, et al. Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): initial results of a randomised phase 1 trial. Lancet 2011;378:1847-57

147.

Makkar RR, Smith RR, Cheng K, et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet 2012;379: 895-904

148.

AutoLogous Human CArdiac-Derived Stem Cell to Treat Ischemic cArdiomyopathy

Review

(ALCADIA). Available from: https:// clinicaltrials.gov/ct2/show/NCT00981006 149.

Kamihata H, Matsubara H, Nishiue T, et al. Implantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines. Circulation 2001;104:1046-52

150.

Takahashi M, Li TS, Suzuki R, et al. Cytokines produced by bone marrow cells can contribute to functional improvement of the infarcted heart by protecting cardiomyocytes from ischemic injury. Am J Physiol Heart Circ Physiol 2006;291: H886-93

151.

Matsumoto R, Omura T, Yoshiyama M, et al. Vascular endothelial growth factor-expressing mesenchymal stem cell transplantation for the treatment of acute myocardial infarction. Arterioscler Thromb Vasc Biol 2005;25:1168-73

152.

Li TS, Cheng K, Malliaras K, et al. Direct comparison of different stem cell types and subpopulations reveals superior paracrine potency and myocardial repair efficacy with cardiosphere-derived cells. J Am Coll Cardiol 2012;59:942-53

153.

Study to Evaluate the Safety of a Single Escalating Dose of ACRX-100 in Adults With Ischemic Heart Failure. Available from: https://clinicaltrials.gov/ct2/show/ NCT01082094

154.

Penn MS, Mendelsohn FO, Schaer GL, et al. An open-label dose escalation study to evaluate the safety of administration of nonviral stromal cell-derived factor1 plasmid to treat symptomatic ischemic heart failure. Circ Res 2013;112:816-25

155.

Hou D, Youssef EA, Brinton TJ, et al. Radiolabeled cell distribution after intramyocardial, intracoronary, and interstitial retrograde coronary venous delivery: implications for current clinical trials. Circulation 2005;112:I150-6

156.

Freyman T, Polin G, Osman H, et al. A quantitative, randomized study evaluating three methods of mesenchymal stem cell delivery following myocardial infarction. Eur Heart J 2006;27:1114-22

157.

Segers VF, Tokunou T, Higgins LJ, et al. Local delivery of protease-resistant stromal cell derived factor-1 for stem cell recruitment after myocardial infarction. Circulation 2007;116:1683-92

158.

Projahn D, Simsekyilmaz S, Singh S, et al. Controlled intramyocardial release of engineered chemokines by biodegradable hydrogels as a treatment approach of

535

Review

Jarrah & Tarzami

Expert Review of Clinical Immunology Downloaded from informahealthcare.com by Nanyang Technological University on 04/25/15 For personal use only.

myocardial infarction. J Cell Mol Med 2014;18:790-800 159.

Kirkwood JM, Strawderman MH, Ernstoff MS, et al. Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group Trial EST 1684. J Clin Oncol 1996;14:7-17

160.

Mocellin S, Pasquali S, Rossi CR, et al. Interferon alpha adjuvant therapy in patients with high-risk melanoma: a systematic review and meta-analysis. J Natl Cancer Inst 2010;102:493-501

161.

Tsuruoka N, Sugiyama M, Tawaragi Y, et al. Inhibition of in vitro angiogenesis by lymphotoxin and interferon-gamma. Biochem Biophys Res Commun 1988;155: 429-35

162.

Wagner TC, Velichko S, Chesney SK, et al. Interferon receptor expression regulates the antiproliferative effects of interferons on cancer cells and solid tumors. Int J Cancer 2004;111:32-42

163.

Lee ME, Miller WL, Edwards BS, et al. Role of endogenous atrial natriuretic factor in acute congestive heart failure. J Clin Invest 1989;84:1962-6

164.

165. 166.

Redfield MM, Aarhus LL, Wright RS, et al. Cardiorenal and neurohumoral function in a canine model of early left ventricular dysfunction. Circulation 1993;87:2016-22

167.

Garcia AG, Wilson RM, Heo J, et al. Interferon-gamma ablation exacerbates myocardial hypertrophy in diastolic heart failure. Am J Physiol Heart Circ Physiol 2012;303:H587-96

168.

Chung ES, Packer M, Lo KH, et al. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial. Circulation 2003;107:3133-40

169.

Louis A, Cleland JG, Crabbe S, et al. Clinical trials update: CAPRICORN, COPERNICUS, MIRACLE, STAF, RITZ-2, RECOVER and RENAISSANCE and cachexia and cholesterol in heart failure. Highlights of the Scientific Sessions of the American College of Cardiology, 2001. Eur J Heart Fail 2001;3:381-7

170.

536

Anker SD, Coats AJ. How to RECOVER from RENAISSANCE? The significance of the results of RECOVER, RENAISSANCE, RENEWAL and ATTACH. Int J Cardiol 2002;86:123-30

171.

Coletta AP, Clark AL, Banarjee P, et al. Clinical trials update: RENEWAL (RENAISSANCE and RECOVER) and ATTACH. Eur J Heart Fail 2002;4:559-61

172.

Mann DL, McMurray JJ, Packer M, et al. Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide

Sinagra G, Gavazzi A. [The ACCLAIM trial]. G Ital Cardiol (Rome) 2008;9:455-60 Munger MA, Johnson B, Amber IJ, et al. Circulating concentrations of proinflammatory cytokines in mild or moderate heart failure secondary to ischemic

Evaluation (RENEWAL). Circulation 2004;109:1594-602

or idiopathic dilated cardiomyopathy. Am J Cardiol 1996;77:723-7 .

An interesting update on several clinical trials using anti TNF-a therapies in patient with failing heart in which all of them failed to show any improvement in cardiac function.

173.

Breedveld FC, Weisman MH, Kavanaugh AF, et al. The PREMIER study: a multicenter, randomized, double-blind clinical trial of combination therapy with adalimumab plus methotrexate versus methotrexate alone or adalimumab alone in patients with early, aggressive rheumatoid arthritis who had not had previous methotrexate treatment. Arthritis Rheum 2006;54:26-37

174.

Keystone E, Heijde D, Mason D Jr, et al. Certolizumab pegol plus methotrexate is significantly more effective than placebo plus methotrexate in active rheumatoid arthritis: findings of a fifty-two-week, phase III, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Arthritis Rheum 2008;58:3319-29

175.

Fleischmann R, Vencovsky J, van Vollenhoven RF, et al. Efficacy and safety of certolizumab pegol monotherapy every 4 weeks in patients with rheumatoid arthritis failing previous disease-modifying antirheumatic therapy: the FAST4WARD study. Ann Rheum Dis 2009;68:805-11

176.

Sandborn WJ, Feagan BG, Marano C, et al. Subcutaneous golimumab maintains clinical response in patients with moderate-to-severe ulcerative colitis. Gastroenterology 2014;146:96-109.e1

Expert Rev. Clin. Immunol. 11(4), (2015)

The duality of chemokines in heart failure.

The failing human heart is a bustling network of intra- and inter-cellular signals and related processes attempting to coordinate a repair mechanism f...
661KB Sizes 5 Downloads 9 Views