Abstract
Inflammation is an important contributor to excess cardiovascular risk and progressive renal injury in people with CKD. Dysregulation of the innate and adaptive immune system is accelerated by CKD and results in increased systemic inflammation, a heightened local vascular inflammatory response leading to accelerated atherosclerosis, and dysfunction of the cardiac and renal endothelium and microcirculation. Understanding and addressing the dysregulated immune system is a promising approach to modifying cardiorenal outcomes in people with CKD. However, targeted pharmacotherapies adopted from trials of non-CKD and cardio-rheumatology populations are only beginning to be developed and tested in human clinical trials. Pharmacotherapies that inhibit activation of the NLRP3 inflammasome and the downstream cytokines IL-1 and IL-6 are the most well-studied. However, most of the available evidence for efficacy is from small clinical trials with inflammatory and cardiorenal biomarker endpoints, rather than cardiovascular event endpoints, or from small CKD subgroups in larger clinical trials. Other pharmacotherapies that have proven beneficial for cardiorenal endpoints in people with CKD have been found to have pleiotropic anti-inflammatory benefits including statins, mineralocorticoid receptor antagonists, SGLT-2 inhibitors, and GLP-1 agonists. Finally, emerging therapies in CKD such as IL-6 inhibition, small-interfering RNA against lipoproteins, AhR inhibitors, and therapies adopted from the renal transplant population including mTOR inhibitors and T regulatory cell promoters may have benefits for cardiorenal and inflammatory endpoints but require further investigation in clinical trials.
Keywords: Chronic kidney disease, immunomodulatory therapies, inflammation, cardiorenal endpoints
Introduction
Chronic kidney disease (CKD) is increasing in prevalence globally1 and is a strong risk factor for atherosclerotic cardiovascular disease2,3. Cardiovascular disease is the most common cause of death among people with CKD4. Worsening CKD severity, including declining estimated glomerular filtration rate (eGFR) and increasing urine albumin to creatinine ratio (UACR), is associated with increased cardiovascular risk5,6. Mechanisms of accelerated cardiovascular disease in CKD are multifactorial and incompletely understood; however, they include the combined effects of metabolic risk factors, inflammation, oxidative stress, renin-angiotensin system activation, vascular and endothelial dysfunction, dialysis, and endocrine dysregulation7,8. Cardiovascular disease management in CKD is challenging, with lower efficacy of statins, and decreased benefit and increased procedural risk of revascularization for obstructive coronary artery disease (CAD)9–11. People with CKD are underrepresented in randomized trials of cardiovascular pharmacotherapies and interventions12. Therefore, therapeutic approaches to address excess cardiovascular disease are frequently adopted from research trials in the non-CKD population.
Systemic inflammation is highly prevalent and multifactorial in people with CKD, with up to 50 percent of patients on dialysis having increased biomarkers of inflammation such as C-reactive protein (CRP)13,14. Systemic inflammation has been consistently linked to the development of atherosclerotic cardiovascular disease15,16, including in CKD17,18. Inflammation is also associated with the initiation and worsening of CKD, further increasing the risk of cardiovascular disease19,20. Elevations of systemic biomarkers of inflammation such as CRP, IL-6, IL-1β, TNF-α, and fibrinogen are common in advanced CKD and dialysis patients14,21, associate with a decline in eGFR22,23, and with increased cardiovascular risk13,17,24,25.
Although there is ample evidence that inflammation has a key role in the development of atherosclerotic cardiovascular disease in CKD, targeted pharmacotherapies to address inflammation and thereby reduce the progression of cardiovascular disease and decrease cardiovascular risk are not yet widely adopted in clinical practice. The purpose of this review is to summarize the key inflammatory mechanisms that may mediate worsening cardiovascular and renal disease in people with CKD, and the available human clinical trial evidence supporting pharmacotherapies that target inflammatory mechanisms both directly and through pleiotropic effects.
Inflammatory mechanisms that promote atherosclerosis among people with CKD
Atherosclerotic plaque is recognized as a local vascular inflammatory and thrombotic response in the context of a systemic inflammatory state and is characterized by deposition of lipids, infiltration of leukocytes, and proliferation of smooth muscle cells in the intima of the arterial wall26,27. The key innate and adaptive immunity pathways that mediate atherosclerosis are accelerated by CKD16. Lipoproteins that are deposited in the intima of the arterial wall undergo oxidative modification by reactive oxygen species (e.g. oxidized low-density lipoprotein, oxLDL), trigger innate immune responses, and subsequently trigger expression of adhesion molecules on endothelial cells such as intracellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1). Monocytes are recruited by endothelial cell surface signals and differentiate into macrophages, which engulf lipoproteins and transform into foam cells, collecting along with cholesterol crystals and apoptotic cellular debris and forming a necrotic core within an atherosclerotic plaque. Adaptive and cellular immunity with local infiltration of T and B cells and self-tolerance aberrations including the development of autoantigens such as oxLDL in the atherosclerotic plaque also play an important but incompletely understood role.16,28
Innate Immunity and the NLRP3 inflammasome
Innate immunity, the initial line of immune defense, is mediated by pattern-recognition receptors (PRRs) on and within immune cells and has a key role in the development of sterile inflammation in atherosclerosis. PRRs recognize antigens from damaged cells, called damage-associated molecular patterns (DAMPs), facilitating the formation and activation of inflammasomes. NOD-like receptors (NLRs) and toll-like receptors (TLRs) have been found to be particularly important in atherosclerosis29,30. NLRs can detect LDL cholesterol and cholesterol crystals and TLRs can detect oxLDL and other ligands released by local tissue damage that are related to chronic atherosclerotic inflammation30–33.
The NOD-like receptor protein 3 (NLRP3) inflammasome is a well-studied mediator of inflammatory atherosclerotic disease. The NLRP3 inflammasome activates caspase-1, which cleaves pro-interleukin-1β (pro-IL-1β) to IL-1β. Through this pathway, NLRP3 has a key role in both atherosclerotic cardiovascular disease34 and the development of chronic kidney injury35,36. Activation of TLRs by DAMPs is a key step in activation of the NLRP3 inflammasome, as they stimulate expression of NLRP3 inflammasome components and pro-IL-1β via nuclear factor-ΚB (NF-κB) 30. IL-1β further stimulates IL-6, which is also associated with atherosclerotic cardiovascular disease risk37 and progression of CKD38. IL-1α, which is expressed constitutively on the cell surface and does not require caspase-1 cleavage for activation, also may mediate cardiovascular disease in CKD. IL-1α has been found to be expressed on monocytes in CKD patients with acute myocardial infarction and is associated with increased risk of recurrent cardiovascular events39. Cytokines such as IL-1 and IL-6 activate endothelial cells, leading to expression of cell adhesion molecules such as VCAM-1 and ICAM-1 and promoting adhesion of other inflammatory cells34. Pro-inflammatory cytokines also induce the production of the acute phase reactant C-reactive protein from the liver40,41.
Role of lipoproteins and cholesterol crystals
There are a variety of atherosclerosis-associated DAMPs that mediate activation of the NLRP3 inflammasome and other pro-inflammatory pathways. Lipoproteins, such as the apolipoprotein ApoC3 have been shown to induce IL-1β release from monocytes via the NLRP3 pathway in conjunction with TLR activation42. Dyslipidemia and modification in lipoproteins are common among people with CKD, including increased LDL and triglyceride levels, reduced high density lipoprotein (HDL) levels, increased ApoB lipoproteins, increased lipoprotein(a), and modification of LDL including carbamylation (cLDL) and oxidation (oxLDL)43,44. Carbamylation of LDL has been shown to be associated with increased urea and myeloperoxidase levels in uremia45. Additionally, systemic inflammation associated with CKD modifies the composition and function of HDL, and therefore may modify the association of HDL with cardiovascular risk. For instance, in the KNOW-CKD study, there was a loss of the protective effect of high HDL in patients with inflammation as measured by elevated CRP, which may be due to dysfunctional HDL46. Finally, CKD is associated with accumulation of proatherogenic cholesterol in a crystalline state (cholesterol crystals)45.
Accumulation of serum uric acid
Chronic kidney disease is a risk factor for hyperuricemia and gout, in part because serum uric acid is primarily excreted by the kidneys. Hyperuricemia has been associated with the development of atherosclerotic CV disease in several meta-analyses of prospective studies47–49. Intracellular uric acid has been shown to induce oxidative stress in pre-clinical studies, triggering down-stream inflammatory signaling pathways including the mitogen-activated protein kinase (MAPK) cascade and NLRP3 inflammasome, as well as influencing macrophage differentiation to foam cells50.
Alterations in the gut microbiome and uremic toxins
CKD alters the gut microbiome and impairs intestinal barrier function. Alterations in the gut microbiome have been linked to increased absorption and reduced clearance of uremic toxins that can cause cardiovascular disease51. For instance, protein-bound toxins such as indoxyl sulfate, p-cresyl sulfate and trimethylamine N-oxide (TMAO) which arise from metabolism of amino acids in the gut by bacteria, have been linked to adverse renal and cardiovascular outcomes51–53. These toxins are linked to increased IL-1B and NF-κB production through an aryl hydrocarbon receptor (AhR) and MAPK pathway54.
Adaptive immunity and the role of T regulatory cells
Following the innate immune response, cells of the adaptive immune system including T cells, B cells and regulatory T cells are recruited in response to antigens on antigen presenting cells (APCs), especially major histocompatibility complex class II (MHC2) on macrophages and dendritic cells. Cells of the adaptive immune system have been shown to have both pathogenic effects, in the case of T helper 1 (TH1) and B-2 cells, and protective effects, in the case of T regulatory (Treg) cells28. CKD and uremia are associated with expansion and dysfunction of the atherogenic T helper cell population and depletion of the Treg population, along with depletion of protective natural anti-oxLDL IgM autoantibodies45.
Therapeutic Approaches: Evidence from Clinical Trials
Targeted immunomodulatory pharmacotherapies and non-targeted pharmacotherapies with pleiotropic anti-inflammatory effects have been studied in people with CKD in clinical trials, showing benefit for inflammatory and cardiorenal biomarker endpoints and less commonly cardiovascular outcome endpoints (Table 1). The pharmacotherapies studied and developed to date are primarily targeted against the innate immune system (Figure 1) however novel therapies are being developed to target the adaptive immune system and other inflammatory pathways.
Table 1:
Targeted and non-targeted immunomodulatory therapies in CKD: Human trial evidence of effect on inflammatory and cardiorenal biomarkers and outcomes
| Immune Pathway | Therapy | Cardiorenal Biomarkers | Inflammatory Biomarkers | Outcomes | Trials |
|---|---|---|---|---|---|
| IL-1 | Canakinumab | ↓ hsCRP | ↓ CV Events | CANTOS59 | |
| Anakinra | ↓ hsCRP, ↓ IL-6, ↓ myeloperoxidase activity | Hung et al55, REDHART56 | |||
| Rilonacept | ↑ FMD | ↓ hsCRP, ↓ NADPH oxidase expression | Nowak et al57, Hung et al58 | ||
| IL-6 | Ziltivekimab | ↓ Lp(a) | ↓ hs-CRP, ↓ fibrinogen | RESCUE62, ZEUS (in progress)63 | |
| Tocilizumab | ↑ salvageable myocardium on MRI, ↓ troponin | ↓ hs-CRP | ASSAIL-MI67, Kleveland et al68 | ||
| Nrf2 | Bardoxolone methyl | ↑ eGFR | ↔ ESRD development, ↔ CV deaths, ↑ heart failure hospitalization & death | BEAM72, BEACON73 | |
| mTOR | Sirolimus | ↓ CIMT | Silva et al76 | ||
| Nontargeted therapies with pleiotropic anti-inflammatory properties | Methotrexate | ↓ eGFR decline | ↔ hsCRP, ↔ IL-6 | ↔ CV events | CIRT (CKD stage 1–3)79,80 |
| Colchicine | ↔ albuminuria | ↓ hsCRP, ↓ NLRP3 | ↔ CV deaths | LoDoCo2 CKD stage 3A subanalyses82,89, Wang et al91 | |
| Other uric acid lowering agents (Allopurinol and Febuxostat) | ↔ eGFR decline | ↔ CRP, ↓ uric acid | ↔ CV events | Goicoechea et al93, CKD-FIX94, PERL95, FEATHER96, FREED97,98 | |
| Statins | ↓ LDL, ↓ eGFR decline | ↓ hsCRP | ↓ CV events, except ↔ in dialysis-dependent in AURORA and 4D | JUPITER CKD subanalysis103, SHARP104, CARE23, AURORA105, 4D106 | |
| MRAs (finerone) | ↓ eGFR decline, ↓ proteinuria | ↓ CV events | FIDELIO-DKD109, FIGARO-DKD110 | ||
| SGLT-2 inhibitors (dapagliflozin, canagliflozin) | ↓ IL-1β, ↓ NLRP3, ↑ BHB, ↓ insulin, ↓ IL-6, ↓ MMP-7, ↓ fibronectin-1, ↓ TNF receptor 1 | ↓ CV events, ↓ adverse renal outcomes | DAPA-CKD113, Kim et al114, CANATA-SU subanalysis115 | ||
| GLP-1 agonists | ↓ eGFR decline, ↓ albuminuria | ↓ CV events | SUSTAIN 6 and LEADER post-hoc analysis116, Zelniker et al metaanalysis117 |
CV: cardiovascular; hsCRP: high-sensitivity C-reactive protein; IL: interleukin; NADPH: nicotinamide adenine dinucleotide diphosphate; FMD: flow-mediated dilation; Lp(a): Lipoprotein (a); Nrf2: nuclear factor erythroid-2-related factor 2; ESRD: end-stage renal disease; mTOR: mammalian target of rapamycin; CIMT: carotid intima media thickness; eGFR: estimated glomerular filtration rate; LDL: low-density lipoprotein; NLRP3: (NOD)-like receptor protein 3; BHB: beta hydroxybutyrate; MMP-7: matrix metalloproteinase 7; MRAs: mineralocorticoid receptor antagonists; TNF: tumor necrosis factor; SGLT-2: Sodium-glucose cotransporter 2; GLP-1: glucagon-like peptide-1
Figure 1:

Innate immunity cellular signaling pathways involved in atherosclerotic cardiovascular disease in people with CKD, and targeted and pleiotropic immunomodulatory therapeutic approaches
*Have not shown a reduction in cardiorenal or inflammatory biomarkers or favorable impact on outcomes in clinical trials
IL: interleukin; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; TLR: Toll-like receptor; MyD88: Myeloid differentiation primary response 88; JAK: Janus kinase; STAT: signal transducer and activator of transcription; NLRP3: (NOD)-like receptor protein 3; ASC: Apoptosis-associated speck-like protein; BHB: beta hydroxybutyrate; SGLT-2: Sodium-glucose cotransporter 2; GLP-1: glucagon-like peptide-1
Targeted immunomodulatory pharmacotherapies
Interleukin-1 (IL-1)
IL-1β inhibition by canakinumab, antagonism of the IL-1 receptor by anakinra and the high affinity trapping of IL-1β and IL-1α by the soluble decoy receptor rilonacept have shown potential benefit to improve cardiovascular biomarkers and outcomes in clinical trials among people with CKD and dialysis-dependent end-stage renal disease (ESRD). These therapies are currently approved and utilized for treatment of autoimmune inflammatory diseases such as rheumatoid arthritis (anakinra) and recurrent idiopathic pericarditis (rilonacept). Anakinra has been shown to reduce high-sensitivity CRP (hsCRP) and IL-6 levels in a small, randomized placebo-controlled trial of 22 dialysis-dependent patients with elevated baseline hsCRP55. In a post-hoc analysis of the Recently Decompensated Heart Failure Anakinra Response Trial (REDHART) study, two weeks of Anakinra was also found to reduce hsCRP and myeloperoxidase activity in 52 patients with eGFR 30–60 ml/min/1.73 m2 and heart failure56. In a further double-blind trial of 42 patients with stage 3–4 CKD, rilonacept reduced hsCRP and endothelial cell nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression, and improved brachial artery flow-mediated dilation (FMD)57. Both anakinra and rilonacept were found to improve HDL anti-oxidant and anti-inflammatory functional properties58.
The strongest evidence to support a beneficial effect of IL-1 blockade on cardiovascular outcomes in CKD comes from a post-hoc analysis of the Canakinumab Anti-Inflammatory Thrombosis Outcome Study (CANTOS) of patients with prior myocardial infarction. In CANTOS, canakinumab significantly reduced the risk of major adverse cardiovascular events by 18% in the 1,875 participants with eGFR 30–60 ml/min/1.73 m2 (HR [95% CI]: 0.82 [0.68–1.00]) and by 14% among those with eGFR > 60 ml/min/1.73 m2 (HR [95% CI]: 0.86 [0.77–0.97]; P-heterogeneity = 0.68).59 A similar pattern of benefit for canakinumab was observed among participants with albuminuria at baseline, although urine albumin-to-creatinine ratio was obtained only for participants with pre-diabetes or diabetes at baseline.59 Canakinumab had minimal effects on eGFR decline, albuminuria progression and the risk of CKD progression.59
Inhibition of IL-1α has not been studied as a therapeutic target for cardiovascular disease in CKD, although it has been found to be a key mediator of inflammation in patients with acute myocardial infarction39. Xilonix is a novel therapy targeting IL-1α that has been studied in non-small lung cancer and colorectal cancer60, as well as in a phase 2 clinical trial following superficial femoral artery revascularization in which it was not associated with reduced cardiovascular events compared to placebo61.
Interleukin-6 (IL-6)
In the recent phase 2 RESCUE trial, the IL-6 monoclonal antibody Ziltivekimab was found to reduce inflammation as measured by hsCRP, fibrinogen, serum amyloid A, haptoglobin, and secretory phospholipase A2 as well as reduce lipoprotein(a) levels62. Evidence of a benefit of IL-6 inhibition on longitudinal cardiovascular outcomes in people with CKD is lacking, however the large clinical trial Ziltivekimab Cardiovascular Outcomes Study (ZEUS) is ongoing63 and Mendelian randomization analyses provide causal evidence for an effect of IL-6 on cardiovascular events among people with CKD64–66. In the non-CKD population, the randomized clinical trial Assessing the effect of anti-IL-6 treatment in MI (ASSAIL-MI) found that the IL-6 receptor antibody tocilizumab improved the proportion of salvageable myocardium on MRI after ST elevation myocardial infarction (STEMI)67. In a separate trial, tocilizumab attenuated the increase in hsCRP and PCI-related troponin release after non-ST elevation myocardial infarction (NSTEMI)68. However, tocilizumab has not been studied in clinical trials of people with CKD, except in renal transplant recipients as described in a separate section. Additionally, tocilizumab has been associated with modest increases in total cholesterol, HDL, LDL and triglyceride levels69; however, it has been shown to have a positive effect on lipoprotein (a) and oxLDL which may imply decreased CV risk despite worsening in some lipid parameters70.
Nuclear factor erythroid-2-related factor 2 agonists
Targeted therapies for the transcription factor nuclear factor erythroid-2-related factor 2 (Nrf2), which is a genetic regulator that promotes antioxidant activity, have been investigated to improve cardiac and renal outcomes. Bardoxolone methyl is an investigational Nrf2 activator which also inhibits NF-κB71. In the Bardoxolone Methyl Treatment: Renal Function in CKD/Type 2 Diabetes (BEAM) phase 2 randomized trial, bardoxolone methyl was found to significantly increase estimated GFR compared to placebo among 227 diabetics with CKD stage 3–472. However, in the Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type 2 Diabetes Mellitus: the Occurrence of Renal Events (BEACON) randomized phase 3 trial of 2185 diabetics with CKD, there was no difference in the primary outcome of development of ESRD or death from cardiovascular causes for people on bardoxolone methyl compared to placebo, and the trial was terminated early because of higher rates of hospitalization or death from heart failure73.
Adaptive Immunity/Regulatory T-cells
Targeted therapies for adaptive immunity are a promising approach but are not well studied in people with CKD. Even among the non-CKD population, the available evidence for modulating adaptive immunity to reduce atherosclerotic cardiovascular disease is primarily in the pre-clinical stage74. Modulation of IL-2 is one potential approach. The mammalian target of rapamycin (mTOR) inhibitor sirolimus blocks the cell-cycle kinase mTOR which has the downstream effect of inhibition of T and B cell activity mediated by IL-275. In a small clinical trial of kidney transplant recipients randomized to tacrolimus/sirolimus vs tacrolimus/mycophenolate, those on sirolimus had a decrease in carotid intima media thickness (CIMT) and had a higher HDL76. The favorable impact of sirolimus on carotid atherosclerosis in this trial was seen despite an association in other clinical trials of mTOR inhibitors with adverse metabolic effects after transplant including new-onset diabetes, elevated cholesterol, and elevated triglycerides77.
Pharmacotherapies with nonspecific or pleiotropic immunomodulatory effect
Methotrexate
Methotrexate non-specifically suppresses systemic inflammation through several mechanisms including inhibition of dihydrofolate reductase leading to increased T cell apoptosis, inhibition of aminoimidazole-4-carboxamide ribonucleotide (AICAR) transformylase (ATIC) leading to adenosine release and cellular binding and suppression of immune reactions, and inhibition of NF-κB78. In the Cardiovascular Inflammation Reduction Trial (CIRT), methotrexate did not reduce cardiovascular events nor did it reduce inflammatory biomarkers such as hsCRP, IL-6, and IL-1β79. However, those randomized to methotrexate had slightly less decline in eGFR compared to placebo, and it was safe in CKD stage 1–380. Methotrexate has not been studied in people with more advanced CKD in a clinical trial, however given the lack of efficacy in CIRT for cardiovascular or inflammatory endpoints, and the potential risk for dose-dependent and CKD-stage-dependent worsening of renal impairment associated with crystal nephropathy81, it is less likely to be a promising anti-inflammatory therapy.
Colchicine
Colchicine is a non-specific anti-inflammatory therapy that has been primarily used in gout and pericarditis. In the Colchicine Cardiovascular Outcomes Trial (COLCOT) and Low-Dose Colchicine 2 (LoDoCo2) trials of patients with recent myocardial infarction and chronic CAD respectively, low-dose colchicine 0.5 mg daily has been found to reduce risk for cardiovascular events and mortality82,83. A new, low-dose formulation of colchicine (0.5 mg daily) was FDA approved in June 2023 to reduce the risk of myocardial infarction (MI), stroke, coronary revascularization, and cardiovascular death in adult patients with established atherosclerotic disease or with multiple risk factors for cardiovascular disease (the standard dose in the United States is 0.6 mg)84,85 and both American and European guidelines endorse the use of colchicine for atherosclerotic cardiovascular disease prevention in select patients86,87. Colchicine achieves high concentrations in leukocytes and has a variety of anti-inflammatory effects including the inhibition of neutrophil migration and adhesion, smooth muscle proliferation, and NLRP3 inflammasome activation which results in reduction of the release of inflammatory markers such as IL-1β and IL-688. In a sub-analysis of the LoDoCo2 trial, colchicine was found to reduce extracellular vesicle NLRP3 levels along with hsCRP89. Low-dose colchicine was well-tolerated in both COLCOT and LoDoCo2, with modestly higher rates of gastrointestinal adverse events, although participants with CKD were excluded from these trials.
The safety profile and efficacy of colchicine for CAD prevention in CKD has not been well studied, with no dedicated clinical trials90. In a study of 190 participants with type 2 diabetes mellitus and moderately increased urine albumin-to-creatinine ratio, low-dose colchicine 0.5 mg daily did not slow the progression of albuminuria but also did not increase the risk of gastrointestinal adverse effects91. Colchicine is contraindicated/not recommended for patients with severe renal impairment85, defined as eGFR <15 ml/min/1.73 m2 in manufacturer guidelines84 and eGFR <30 ml/min/1.73 m2 in the 2023 American guidelines for chronic CAD86. Colchicine steady-state serum levels may exceed accepted safe limits and increase risk for adverse effects in the setting of renal impairment, based on pharmacodynamic modeling studies85. In a subgroup analysis of the LoDoCo2 trial, of patients with stage 3A CKD which included only 148 patients, there was no association of colchicine with cardiovascular events (HR 1.18 [95% CI 0.53–2.63]82. There is some evidence that colchicine may be effective for renal endpoints in moderate CKD, including a nested case-control study which found that colchicine use in stage 3 to 4 CKD was associated with reduced risk of CKD progression92.
Uric Acid Lowering Therapies
Hyperuricemia has been linked to CKD, CV disease and inflammation, and uric acid lowering therapies for gout including allopurinol and febuxostat have been studied as potential therapies to reduce cardiovascular and renal risk50. In a small non-placebo-controlled trial of 113 participants with CKD stage 3 and greater randomized to allopurinol or continuation of their usual therapy, allopurinol was associated with decline in serum uric acid and CRP, fewer cardiovascular events, and slower decline in eGFR93. However, subsequent larger and placebo-controlled trials have not found beneficial effects on cardiovascular and renal outcomes. In the Controlled Trial of Slowing of Kidney Disease Progression from the Inhibition of Xanthine Oxidase (CKD-FIX), which randomized 369 patients to allopurinol or placebo, there was no difference in the primary outcome of change in eGFR at 104 weeks, or difference in secondary outcomes including development of ESRD or cardiovascular events94. The Preventing Early Renal Loss in Diabetes (PERL) trial randomized 267 patients with type 1 diabetes and early to moderate stage CKD to allopurinol or placebo, and did not find a difference in the primary outcome of change in iohexol-based GFR after 3 years, or secondary outcomes of creatine doubling/ESRD or cardiovascular events95. In the Febuxostat Versus Placebo Randomized Controlled Trial With Hyperuricemia Complicated by Chronic Kidney Disease Stage 3 (FEATHER), which randomized 467 patients with stage 3 CKD to febuxostat or placebo, there was no difference in the primary outcome of slope of eGFR change after 108 weeks96. Finally, in the Febuxostat for Cerebral and Cardiorenovascular Events Prevention Study (FREED), which randomized 1070 patients to febuxostat or usual care (which could include allopurinol), the primary composite event of cerebral, cardiovascular, and renal events was lower in the febuxostat group. However, the difference was driven by development of renal impairment, which included the development or worsening of albuminuria, and there was no difference in the slope of eGFR change or in cardiovascular events97. Additionally, in a post-hoc analysis there was no difference in hsCRP levels after febuxostat treatment in the FREED trial98. In summary, the combined clinical trial evidence of uric acid lowering therapies in people with CKD does not support a benefit for cardiovascular or renal outcomes.
Statins
Statins are among the most well-studied therapies to reduce cardiovascular disease risk, including in people with CKD99, and have been found to reduce systemic inflammation100. Some, but not all, statins have been found to have pleiotropic anti-inflammatory effects, including through lipid-lowering and inhibition of the NLRP3 inflammasome and Toll-like receptors30,101. In a meta-analysis of 25 studies in patients with CKD, statin therapy decreased CRP by −2.06 mg/L (95% CI −2.85 to −1.27)100. In the Justification for the Use of Statin in Prevention: An Intervention Trial Evaluating Rosuvastatin (JUPITER) trial, which enrolled healthy individuals with elevated hsCRP, rosuvastatin lowered risk of cardiovascular events and the hsCRP concentration102. In a sub-analysis of the JUPITER trial, in patients with eGFR <60 ml/min/1.73 m2, rosuvastatin also lowered risk of cardiovascular events and the hsCRP concentration103. The Study of Heart and Renal Protection (SHARP) trial found that simvastatin plus ezetimibe reduced risk of cardiovascular events in both dialysis-dependent and non-dialysis dependent CKD104. In a sub-analysis of participants with eGFR <60 in the Cholesterol and Recurrent Events (CARE) trial of pravastatin therapy in patients with history of myocardial infarction, higher CRP was associated with renal function decline and pravastatin was associated with prevention of renal function decline23. Conversely, in the A Study to Evaluate the Use of Rosuvastatin in Subjects on Regular Hemodialysis: An Assessment of Survival and Cardiovascular Event (AURORA) and Die Deutsche Diabetes Dialyse (4D) randomized trials of hemodialysis patients, rosuvastatin and atorvastatin lowered LDL cholesterol and either lowered or stabilized CRP compared to placebo but did not reduce the risk for cardiovascular events105–107. In a post-hoc analysis of 4D, although baseline inflammation as measured by CRP was high, and higher CRP was predictive of cardiovascular events, atorvastatin did not have an influence on outcomes regardless of quartile of baseline CRP107. In summary, based on current trial evidence, statins have pleiotropic anti-inflammatory benefits in patients with both dialysis-dependent and non-dialysis dependent CKD, however the benefit on CV outcomes is attenuated or absent in patients on dialysis.
Mineralocorticoid Receptor Antagonists
Mineralocorticoid receptor antagonists have been found to reduce proteinuria,108 and reduce CKD progression and cardiovascular events in the Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease (FIDELIO-DKD) and Finerenone in Reducing Cardiovascular Mortality and Morbidity in Diabetic Kidney Disease (FIGARO-DKD) randomized trials.109,110 Mineralocorticoid activation and hyperaldosteronism are associated with adverse myocardial remodeling secondary to fibrosis, mitochondrial dysfunction, and dysregulation of beta-adrenergic receptor and insulin signaling111. In pre-clinical studies, mineralocorticoid receptor activation has also been linked to increases in NADPH oxidase activity and reactive oxygen species that mediate pro-inflammatory cytokines such as TGF- β and IL-1β108,112. Therefore, it is plausible that the benefit of MR blockade on CKD progression and cardiovascular events goes beyond direct effects on myocardial structure and function and is partially mediated by anti-inflammatory mechanisms. However clinical trial evidence is not currently available to demonstrate this hypothesis.
Sodium-glucose cotransporter 2 (SGLT2) inhibitors
Sodium-glucose cotransporter 2 (SGLT2) inhibitors increase urinary glucose excretion and reduce blood glucose, but also have been found to reduce cardiovascular risk which may be mediated by pleiotropic anti-inflammatory effects. In the Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease (DAPA-CKD) trial of CKD stage 2–4, dapagliflozin was associated with lower risk of adverse renal outcomes and cardiovascular events113. In a small randomized trial of diabetics, the SGLT2-inhibitor empagliflozin was found to reduce activation of the NLRP3 inflammasome and reduce IL-1β secretion in macrophages, which was in part found to be mediated by an increase in β-hydroxybutyrate (BHB) and a decrease in insulin.114 In a sub-analysis of blood samples from the Canagliflozin Treatment And Trial Analysis versus Sulfonylurea (CANTATA-SU) trial of people with diabetic kidney disease, the SGLT2-inhibitor canagliflozin was found to have potential anti-inflammatory and anti-fibrotic effects including decrease in IL-6, fibronectin 1, TNF receptor 1, and matrix metalloproteinase 7 (MMP-7) levels compared with glimepiride115.
Glucagon-like peptide-1 receptor agonists
In a post hoc analysis of the trials of the glucagon-like peptide-1 (GLP-1) receptor agonists semaglutide and liraglutide, people with diabetes and CKD on GLP-1 receptor agonists had lower albuminuria and less decline in eGFR in addition to their previously described beneficial effects on cardiovascular events116. Additionally, in a metanalysis of 5 trials of GLP-1 receptor agonists (Lixisenatide – ELIXA; Liraglutide – LEADER; Semaglutide – SUSTAIN-6; Exenatide – EXSCEL; Albiglutide – HARMONY) compared to 3 trials of SGLT2 inhibitors (Empaglifloxin - EMPA-REG OUTCOME; Canagliflozin – CANVAS; Dapagliflozin – DECLARE TIMI 58), both had similar benefit for reduction of cardiovascular events and also reduced development of macroalbuminuria. However, only SGLT2 inhibitors reduced risk of worsening eGFR, renal death, or ESRD117. There is limited prospective human data on whether GLP-1 agonists modify inflammatory biomarkers in people with CKD. However, in the non-CKD population there is extensive human prospective study evidence of the anti-inflammatory effect including inhibition of the expression of NF-κB, IL-1β and TLRs and reduction in hsCRP and oxidative stress118,119. These anti-inflammatory effects are likely to translate to people with CKD.
Role of Inflammation in Renal transplant Recipients
Renal transplantation is associated with increased activation of the immune system120. Furthermore, renal transplantation is associated with a reduction of cardiovascular risk, although the absolute risk remains high even after transplant121,122. Inflammatory markers are elevated among renal transplant recipients and associated with increased risk of graft loss and mortality123. Targeting innate and adaptive immunity after transplant may provide beneficial effects against inflammation, reduce risk for allograft injury and rejection, and improve cardiorenal outcomes. However, most trials of immunomodulatory therapies in renal transplant recipients have focused on antibody mediated rejection and allograft loss and have not included cardiovascular endpoints.
Among renal transplant recipients in the Assessment of Lescol in Renal Transplantation (ALERT) trial who were randomized to Fluvastatin, IL-6 and hsCRP were associated with increased risk of cardiovascular events and all-cause mortality in a post-hoc analysis124. Fluvastatin was associated with a reduction in cardiovascular death and myocardial infarction, which was a secondary outcome125.
IL-6 inhibition has shown promise for improving renal transplant outcome; small studies have targeted IL-6 inhibition through Tocilizumab (IL-6 receptor antibody) and Clazakizumab (direct IL-6 antibody) and demonstrated reduced rates of acute rejection and improvement in graft survival, renal function and patient survival126. However, human trials have not been performed to assess the efficacy of targeting IL-6 to modify cardiovascular endpoints in the post-transplant population. Additionally, IL-6 receptor inhibition by Tocilizumab has been associated with increased IL-6 levels127 and potential rebound inflammation when discontinued126, and therefore direct inhibition of IL-6 with Clazakizumab or Ziltivekimab may be more promising to improve cardiorenal outcomes.
The mTOR pathway and adaptive immunity may be another possible target for patients after renal transplant. Sirolimus use was associated with a decrease in carotid intima media thickness (CIMT) and an increase in HDL in a small clinical trial comparing tacrolimus/sirolimus vs tacrolimus/mycophenolate76. However, in the ELEVATE trial of conversion from a calcineurin inhibitor to everolimus, there was no change in eGFR or left ventricular mass index, and acute rejection rate was higher compared to patients remaining on a calcineurin inhibitor128.
T regulatory cell stimulation is another potential therapeutic target, which has shown initial promise for managing acute rejection and maintaining tolerance of the transplanted organ129. Because T regulatory cells are also potentially protective in the inflammatory pathways involved with atherosclerosis, they may also improve cardiovascular outcomes, however this has not been explored in a human trial.
Future Directions
NLRP3 inflammasome mediated IL-1 and IL-6 cytokine release is a promising pharmacotherapeutic pathway to improve renal and cardiovascular outcomes. The phase 3 randomized clinical trial ZEUS which began enrollment in 2021 and will complete in 2025, has the objective of comparing IL-6 inhibition with ziltivekiumab to placebo for cardiorenal outcomes in people with CKD63. Larger trials of therapies targeting IL-1, the IL-1 receptor, the NLRP3 inflammasome and IL-1α in people with CKD with assessment of cardiorenal and inflammatory outcomes are needed. Targeted lipoprotein therapies have also shown promise in the non-CKD population. The small-interfering RNA Olpasiran has been found to reduce lipoprotein(a) levels in the Olpasiran Trials of Cardiovascular Events and Lipoprotein[a] Reduction–Dose Finding Study (OCEAN(a) DOSE) trial130, and whether it improves CV outcomes is currently being investigated in a phase 3 trial. Lipoprotein(a) is linked to arterial wall and systemic inflammation, by acting as a DAMP that can trigger an inflammatory response along with oxLDL and through interactions with IL-6131. People with CKD, and especially dialysis dependent ESRD, may especially benefit from novel lipid modifying therapies because they have been found to have a higher prevalence of elevated lipoprotein(a) along with other lipid abnormalities43. However, patients with CKD stage 4 and greater are excluded from the OCEAN(a) trials130 and thus further trials of people with CKD will need to be designed.
There are also promising targeted anti-inflammatory therapies to address cardiorenal disease that are still in the pre-clinical phase. Uremic toxins such as the gut metabolite indoxyl sulfate have been linked to atherosclerosis and impairment in microcirculatory angiogenesis through the aryl hydrocarbon receptor (AhR)53,132. AhR has been linked to atherosclerosis through a role in foam cell formation, upregulation of ICAM-1 and monocyte chemoattractant protein 1 (MCP-1) and activation of NF-κB with release of IL-1β54,132–134. AhR inhibitors are being studied in non-cardiovascular applications such as cancer chemotherapy,135 and could potentially be adapted for the treatment of cardiovascular disease in people with CKD. Other therapies that target adaptive immunity including IL-2 modulation, mTOR inhibitors, and T regulatory cells are potential future areas of investigation, which could be adapted from pre-clinical models and the renal transplant recipient population into human clinical trials of people with CKD.
Finally, more specific biomarkers of cardiovascular and renal inflammation are needed to serve as intermediate endpoints in initial therapeutic trials prior to larger studies which are powered for cardiovascular outcome endpoints. Trials of immunomodulatory therapy in CKD have relied on non-specific systemic biomarkers of inflammation such as hsCRP and IL-6. Advanced imaging approaches are especially promising. These included targeted imaging of vascular inflammation using radiotracer-labeled fluorodeoxyglucose (FDG) and DOTATATE positron emission tomography (PET)136,137, assessment of coronary vascular function with myocardial perfusion PET imaging138, and assessment of coronary artery inflammation from measurement of perivascular fat attenuation with cardiac CT139.
Conclusions
Inflammation is a key mediator of worsening cardiovascular and renal disease in people with CKD, however targeted pharmacotherapies that address inflammatory mechanisms such as IL-1, IL-6 and the NLRP3 inflammasome are only beginning to be developed and studied in clinical trials assessing longitudinal cardiovascular and renal outcomes. Clinical trials have increasingly incorporated assessment of inflammatory endpoints in people with CKD23,57,58,63,103, and therapies with proven cardiorenal benefit such as statins, SGLT-2 inhibitors, GLP-1 agonists and MRAs have been recognized to have pleiotropic anti-inflammatory effects, even though they are not directly targeted against the immune system23,103,114,115. As the cardio-nephrology community is increasingly recognizing the importance of the innate and adaptive immune system in the development of atherosclerosis in CKD, much can be learned from the approach to studying cardiovascular disease in people with rheumatologic systemic inflammatory disorders. It will be important to prioritize pharmacotherapies that target entwined inflammatory and cardiorenal mechanisms. The safety of immunomodulatory therapies in advanced CKD and dialysis-dependent patients, especially the potential increased risk of infection, will need to be established in clinical trials. Incorporating more specific measures of cardiovascular and renal microcirculatory inflammation and dysfunction, which serve as intermediate endpoints in individual patients and smaller trials, will be particularly important. These include promising advanced imaging measures such as vascular inflammation and myocardial perfusion PET, myocardial and pericardial tissue inflammation characterization on MRI, and perivascular adipose tissue-based coronary inflammation assessment on CT. Although there may be challenges ahead in the adoption of immunomodulatory therapies to CKD, early data from clinical trials has demonstrated that there is great promise to the therapeutic targeting of inflammation to address the excess atherosclerotic CV risk in people with kidney disease.
Funding Source:
DMH is supported by a National Institutes of Health (NIH) T32 grant [grant number 5T32HL094301–12]. LFB is supported by a NIH K23 grant [HL150311], ASN/KidneyCure Carl W. Gottschalk Research Scholar Grant and the BWH Khoury Innovation Fund. BW is supported by an American Heart Association Career Development Award [grant number 21CDA851511], NIH/National Heart Lung and Blood Institute K23 grant [HL159276] and the American Society of Nuclear Cardiology IANC Research award.
Disclosures:
LFB reports consulting fees from Kiniksa Pharmaceuticals, LLC. AC reports grants from Bristol-Myers Squibb, ViroPharma, Inc., IQVIA RDS Inc., CSL Behring LLC, Natera Inc., Amgen Inc., Hansa Biopharm AB, Sanofi US Services Inc., Allovir Inc. and personal consulting fees from Allovir, eGenesis, Immucor, Natera consulting and Shire. RB reports research support from Amgen Inc, Novartis Inc, Beren Inc. BW reports /scientific advisory board fees from Novo Nordisk, Kiniksa Pharmaceuticals, and Horizon Therapeutics.
Abbreviations:
- CKD
chronic kidney disease
- ESRD
end-stage renal disease
- eGFR
estimated glomerular filtration rate
- UACR
urine albumin to creatine ratio
- CAD
coronary artery disease
- CRP
C-reactive protein
- hsCRP
high-sensitivity CRP
- IL-6
Interleukin-6
- IL-1β
Interleukin-1 beta
- TNF-α
Tumor necrosis factor alpha
- NF-κB
nuclear factor kappa-light-chain-enhancer of activated B cells
- LDL
low-density lipoprotein
- oxLDL
oxidized LDL
- cLDL
carbamylated LDL
- HDL
high-density lipoprotein
- ICAM-1
intracellular adhesion molecule 1
- VCAM-1
vascular cell adhesion molecule 1
- PRRs
Pattern-recognition receptors
- DAMPs
damage-associated molecular patterns
- NLRs
NOD-like receptors
- TLRs
Toll-like receptors
- NLRP3
(NOD)-like receptor protein 3
- TMAO
trimethylamine N-oxide
- AhR
aryl hydrocarbon receptor
- MAPK
mitogen-activated protein kinase
- APCs
antigen presenting cells
- MHC-2
major histocompatibility complex class II
- TH1
T helper
- Treg
T regulatory cell
- NADPH
nicotinamide adenine dinucleotide phosphate
- FMD
flow-mediated dilation
- STEMI
ST elevation myocardial infarction
- NSTEMI
non-ST elevation myocardial infarction
- Nrf2
nuclear factor erythroid-2-related factor 2
- mTOR
mammalian target of rapamycin
- CIMT
carotid intima media thickness
- ATIC
aminoimidazole-4-carboxamide ribonucleotide transformylase
- MMP-7
matrix metalloproteinase 7
- MCP-1
monocyte chemoattractant protein 1
Footnotes
Conflict of Interest DMH reports no disclosures.
References
- 1.James SL, Abate D, Abate KH, Abay SM, Abbafati C, Abbasi N, Abbastabar H, Abd-Allah F, Abdela J, Abdelalim A, et al. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018;392:1789–1858. doi: 10.1016/S0140-6736(18)32279-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Gansevoort RT, Correa-Rotter R, Hemmelgarn BR, Jafar TH, Heerspink HJL, Mann JF, Matsushita K, Wen CP. Chronic kidney disease and cardiovascular risk: epidemiology, mechanisms, and prevention. Lancet 2013;382:339–352. doi: 10.1016/s0140-6736(13)60595-4 [DOI] [PubMed] [Google Scholar]
- 3.Go AS, Chertow GM, Fan D, E MC, Hsu CY. Chronic Kidney Disease and the Risks of Death, Cardiovascular Events, and Hospitalization. N Engl J Med 2004;351:1296–1305. [DOI] [PubMed] [Google Scholar]
- 4.Thompson S, James M, Wiebe N, Hemmelgarn B, Manns B, Klarenbach S, Tonelli M, Alberta Kidney Disease N. Cause of Death in Patients with Reduced Kidney Function. J Am Soc Nephrol 2015;26:2504–2511. doi: 10.1681/ASN.2014070714 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Chronic Kidney Disease Prognosis C, Matsushita K, van der Velde M, Astor BC, Woodward M, Levey AS, de Jong PE, Coresh J, Gansevoort RT. Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis. Lancet 2010;375:2073–2081. doi: 10.1016/S0140-6736(10)60674-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Matsushita K, Coresh J, Sang Y, Chalmers J, Fox C, Guallar E, Jafar T, Jassal SK, Landman GW, Muntner P, et al. Estimated glomerular filtration rate and albuminuria for prediction of cardiovascular outcomes: a collaborative meta-analysis of individual participant data. Lancet Diabetes Endocrinol 2015;3:514–525. doi: 10.1016/s2213-8587(15)00040-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Schiffrin EL, Lipman ML, Mann JF. Chronic kidney disease: effects on the cardiovascular system. Circulation 2007;116:85–97. doi: 10.1161/CIRCULATIONAHA.106.678342 [DOI] [PubMed] [Google Scholar]
- 8.Jankowski J, Floege J, Fliser D, Bohm M, Marx N. Cardiovascular Disease in Chronic Kidney Disease: Pathophysiological Insights and Therapeutic Options. Circulation 2021;143:1157–1172. doi: 10.1161/CIRCULATIONAHA.120.050686 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sarnak MJ, Amann K, Bangalore S, Cavalcante JL, Charytan DM, Craig JC, Gill JS, Hlatky MA, Jardine AG, Landmesser U, et al. Chronic Kidney Disease and Coronary Artery Disease: JACC State-of-the-Art Review. J Am Coll Cardiol 2019;74:1823–1838. doi: 10.1016/j.jacc.2019.08.1017 [DOI] [PubMed] [Google Scholar]
- 10.Bangalore S, Maron DJ, O’Brien SM, Fleg JL, Kretov EI, Briguori C, Kaul U, Reynolds HR, Mazurek T, Sidhu MS, et al. Management of Coronary Disease in Patients with Advanced Kidney Disease. N Engl J Med 2020;382:1608–1618. doi: 10.1056/NEJMoa1915925 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.KDIGO. KDIGO Clinical Practice Guideline for Lipid Management in Chronic Kidney Disease. Kidney Int November 2013;3. [Google Scholar]
- 12.Konstantinidis ING, Yacoub R, Saha A, Simoes P, Parikh CR, Coca SG. Representation of Patients With Kidney Disease in Trials of Cardiovascular Interventions: An Updated Systematic Review. JAMA Intern Med 2016;176:121–124. doi: 10.1001/jamainternmed.2015.6508 [DOI] [PubMed] [Google Scholar]
- 13.Zimmermann J, Herrlinger S, Pruy A, Metzger T, Wanner C. Inflammation enhances cardiovascular risk and mortality in hemodialysis patients. Kidney Int 1999;55:648–658. doi: 10.1046/j.1523-1755.1999.00273.x [DOI] [PubMed] [Google Scholar]
- 14.Jofré R, Rodriguez-Benitez P, López-Gómez JM, Pérez-Garcia R. Inflammatory Syndrome in Patients on Hemodialysis. JASN 2006;17:S274–S280. doi: 10.1681/asn.2006080926 [DOI] [PubMed] [Google Scholar]
- 15.Ross R. Atherosclerosis - An Inflammatory Disease. NEJM 1999;340:115–126. [DOI] [PubMed] [Google Scholar]
- 16.Gistera A, Hansson GK. The immunology of atherosclerosis. Nat Rev Nephrol 2017;13:368–380. doi: 10.1038/nrneph.2017.51 [DOI] [PubMed] [Google Scholar]
- 17.Wanner C, Zimmermann J, Schwedler S, Metzger T. Inflammation and cardiovascular risk in dialysis patients. Kidney Int Suppl 2002:99–102. doi: 10.1046/j.1523-1755.61.s80.18.x [DOI] [PubMed]
- 18.Wanner C, Amann K, Shoji T. The heart and vascular system in dialysis. The Lancet 2016;388:276–284. doi: 10.1016/s0140-6736(16)30508-6 [DOI] [PubMed] [Google Scholar]
- 19.Tonelli M, Sacks F, Pfeffer M, Jhangri GS, Curhan G, Cholesterol, Recurrent Events Trial I. Biomarkers of inflammation and progression of chronic kidney disease. Kidney Int 2005;68:237–245. doi: 10.1111/j.1523-1755.2005.00398.x [DOI] [PubMed] [Google Scholar]
- 20.Amdur RL, Feldman HI, Gupta J, Yang W, Kanetsky P, Shlipak M, Rahman M, Lash JP, Townsend RR, Ojo A, et al. Inflammation and Progression of CKD: The CRIC Study. Clin J Am Soc Nephrol 2016;11:1546–1556. doi: 10.2215/CJN.13121215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gupta J, Mitra N, Kanetsky PA, Devaney J, Wing MR, Reilly M, Shah VO, Balakrishnan VS, Guzman NJ, Girndt M, et al. Association between Albuminuria, Kidney Function, and Inflammatory Biomarker Profile in CKD in CRIC. Clinical Journal of the American Society of Nephrology 2012;7:1938–1946. doi: 10.2215/cjn.03500412 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Amdur RL, Feldman HI, Gupta J, Yang W, Kanetsky P, Shlipak M, Rahman M, Lash JP, Townsend RR, Ojo A, et al. Inflammation and Progression of CKD: The CRIC Study. Clinical Journal of the American Society of Nephrology 2016;11:1546–1556. doi: 10.2215/cjn.13121215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Tonelli M, Sacks F, Pfeffer M, Jhangri GS, Curhan G. Biomarkers of inflammation and progression of chronickidney disease. Kidney International 2005;68:237–245. doi: 10.1111/j.1523-1755.2005.00398.x [DOI] [PubMed] [Google Scholar]
- 24.Yeun JY, Levine RA, Mantadilok V, Kaysen GA. C-Reactive protein predicts all-cause and cardiovascular mortality in hemodialysis patients. Am J Kidney Dis 2000;35:469–476. doi: 10.1016/s0272-6386(00)70200-9 [DOI] [PubMed] [Google Scholar]
- 25.Menon V, Wang X, Greene T, Beck GJ, Kusek JW, Marcovina SM, Levey AS, Sarnak MJ. Relationship between C-reactive protein, albumin, and cardiovascular disease in patients with chronic kidney disease. Am J Kidney Dis 2003;42:44–52. doi: 10.1016/s0272-6386(03)00407-4 [DOI] [PubMed] [Google Scholar]
- 26.Libby P, Ridker PM, Maseri A. Inflammation and Atherosclerosis. Circulation 2002;105:1135–1143. doi: doi: 10.1161/hc0902.104353 [DOI] [PubMed] [Google Scholar]
- 27.Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med 2005;352:1685–1695. doi: 10.1056/NEJMra043430 [DOI] [PubMed] [Google Scholar]
- 28.Lahoute C, Herbin O, Mallat Z, Tedgui A. Adaptive immunity in atherosclerosis: mechanisms and future therapeutic targets. Nature Reviews Cardiology 2011;8:348–358. doi: 10.1038/nrcardio.2011.62 [DOI] [PubMed] [Google Scholar]
- 29.Speer T, Dimmeler S, Schunk SJ, Fliser D, Ridker PM. Targeting innate immunity-driven inflammation in CKD and cardiovascular disease. Nat Rev Nephrol 2022;18:762–778. doi: 10.1038/s41581-022-00621-9 [DOI] [PubMed] [Google Scholar]
- 30.Koushki K, Shahbaz SK, Mashayekhi K, Sadeghi M, Zayeri ZD, Taba MY, Banach M, Al-Rasadi K, Johnston TP, Sahebkar A. Anti-inflammatory Action of Statins in Cardiovascular Disease: the Role of Inflammasome and Toll-Like Receptor Pathways. Clin Rev Allergy Immunol 2021;60:175–199. doi: 10.1007/s12016-020-08791-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to damage. Nature Reviews Immunology 2010;10:826–837. doi: 10.1038/nri2873 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nuñez G, Schnurr M, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010;464:1357–1361. doi: 10.1038/nature08938 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Xu XH, Shah PK, Faure E, Equils O, Thomas L, Fishbein MC, Luthringer D, Xu X-P, Rajavashisth TB, Yano J, et al. Toll-Like Receptor-4 Is Expressed by Macrophages in Murine and Human Lipid-Rich Atherosclerotic Plaques and Upregulated by Oxidized LDL. Circulation 2001;104:3103–3108. doi: doi: 10.1161/hc5001.100631 [DOI] [PubMed] [Google Scholar]
- 34.Libby P Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J Am Coll Cardiol 2017;70:2278–2289. doi: 10.1016/j.jacc.2017.09.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Komada T, Muruve DA. The role of inflammasomes in kidney disease. Nat Rev Nephrol 2019;15:501–520. doi: 10.1038/s41581-019-0158-z [DOI] [PubMed] [Google Scholar]
- 36.Hutton HL, Ooi JD, Holdsworth SR, Kitching AR. The NLRP3 inflammasome in kidney disease and autoimmunity. Nephrology (Carlton) 2016;21:736–744. doi: 10.1111/nep.12785 [DOI] [PubMed] [Google Scholar]
- 37.Batra G, Ghukasyan Lakic T, Lindbäck J, Held C, White HD, Stewart RAH, Koenig W, Cannon CP, Budaj A, Hagström E, et al. Interleukin 6 and Cardiovascular Outcomes in Patients With Chronic Kidney Disease and Chronic Coronary Syndrome. JAMA Cardiol 2021;6. doi: 10.1001/jamacardio.2021.3079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kreiner FF, Kraaijenhof JM, von Herrath M, Hovingh GKK, von Scholten BJ. Interleukin 6 in diabetes, chronic kidney disease, and cardiovascular disease: mechanisms and therapeutic perspectives. Expert Review of Clinical Immunology 2022;18:377–389. doi: 10.1080/1744666x.2022.2045952 [DOI] [PubMed] [Google Scholar]
- 39.Schunk SJ, Triem S, Schmit D, Zewinger S, Sarakpi T, Becker E, Hütter G, Wrublewsky S, Küting F, Hohl M, et al. Interleukin-1α Is a Central Regulator of Leukocyte-Endothelial Adhesion in Myocardial Infarction and in Chronic Kidney Disease. Circulation 2021;144:893–908. doi: 10.1161/circulationaha.121.053547 [DOI] [PubMed] [Google Scholar]
- 40.Zhang D, Jiang SL, Rzewnicki D, Samols D, Kushner I. The effect of interleukin-1 on C-reactive protein expression in Hep3B cells is exerted at the transcriptional level. Biochem J 1995;310 ( Pt 1):143–148. doi: 10.1042/bj3100143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ridker PM. From C-Reactive Protein to Interleukin-6 to Interleukin-1. Circ Res 2016;118:145–156. doi: doi: 10.1161/CIRCRESAHA.115.306656 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zewinger S, Reiser J, Jankowski V, Alansary D, Hahm E, Triem S, Klug M, Schunk SJ, Schmit D, Kramann R, et al. Apolipoprotein C3 induces inflammation and organ damage by alternative inflammasome activation. Nat Immunol 2020;21:30–41. doi: 10.1038/s41590-019-0548-1 [DOI] [PubMed] [Google Scholar]
- 43.Florens N, Calzada C, Lyasko E, Juillard L, Soulage CO. Modified Lipids and Lipoproteins in Chronic Kidney Disease: A New Class of Uremic Toxins. Toxins (Basel) 2016;8. doi: 10.3390/toxins8120376 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Speer T, Owala FO, Holy EW, Zewinger S, Frenzel FL, Stähli BE, Razavi M, Triem S, Cvija H, Rohrer L, et al. Carbamylated low-density lipoprotein induces endothelial dysfunction. European Heart Journal 2014;35:3021–3032. doi: 10.1093/eurheartj/ehu111 [DOI] [PubMed] [Google Scholar]
- 45.Swaminathan S, Shah SV. Novel inflammatory mechanisms of accelerated atherosclerosis in kidney disease. Kidney International 2011;80:453–463. doi: 10.1038/ki.2011.178 [DOI] [PubMed] [Google Scholar]
- 46.Kim JY, Park JT, Kim HW, Chang TI, Kang EW, Ahn C, Oh KH, Lee J, Chung W, Kim YS, et al. Inflammation Alters Relationship Between High‐Density Lipoprotein Cholesterol and Cardiovascular Risk in Patients With Chronic Kidney Disease: Results From KNOW‐CKD. JAHA 2021;10:e021731. doi: doi: 10.1161/JAHA.120.021731 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zuo T, Liu X, Jiang L, Mao S, Yin X, Guo L. Hyperuricemia and coronary heart disease mortality: a meta-analysis of prospective cohort studies. BMC Cardiovasc Disord 2016;16:207. doi: 10.1186/s12872-016-0379-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Li M, Hu X, Fan Y, Li K, Zhang X, Hou W, Tang Z. Hyperuricemia and the risk for coronary heart disease morbidity and mortality a systematic review and dose-response meta-analysis. Sci Rep 2016;6:19520. doi: 10.1038/srep19520 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhao G, Huang L, Song M, Song Y. Baseline serum uric acid level as a predictor of cardiovascular disease related mortality and all-cause mortality: a meta-analysis of prospective studies. Atherosclerosis 2013;231:61–68. doi: 10.1016/j.atherosclerosis.2013.08.023 [DOI] [PubMed] [Google Scholar]
- 50.Kimura Y, Tsukui D, Kono H. Uric Acid in Inflammation and the Pathogenesis of Atherosclerosis. Int J Mol Sci 2021;22. doi: 10.3390/ijms222212394 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Jovanovich A, Isakova T, Stubbs J. Microbiome and Cardiovascular Disease in CKD. Clin J Am Soc Nephrol 2018;13:1598–1604. doi: 10.2215/cjn.12691117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Vanholder R, Schepers E, Pletinck A, Nagler EV, Glorieux G. The Uremic Toxicity of Indoxyl Sulfate and p-Cresyl Sulfate: A Systematic Review. JASN 2014;25:1897–1907. doi: 10.1681/asn.2013101062 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Arinze NV, Yin W, Lotfollahzadeh S, Napoleon MA, Richards S, Walker JA, Belghasem M, Ravid JD, Hassan Kamel M, Whelan SA, et al. Tryptophan metabolites suppress the Wnt pathway and promote adverse limb events in chronic kidney disease. J Clin Invest 2022;132. doi: 10.1172/jci142260 [DOI] [PMC free article] [PubMed]
- 54.Wakamatsu T, Yamamoto S, Ito T, Sato Y, Matsuo K, Takahashi Y, Kaneko Y, Goto S, Kazama JJ, Gejyo F, et al. Indoxyl Sulfate Promotes Macrophage IL-1β Production by Activating Aryl Hydrocarbon Receptor/NF-κ/MAPK Cascades, but the NLRP3 inflammasome Was Not Activated. Toxins (Basel) 2018;10. doi: 10.3390/toxins10030124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hung AM, Ellis CD, Shintani A, Booker C, Ikizler TA. IL-1beta receptor antagonist reduces inflammation in hemodialysis patients. J Am Soc Nephrol 2011;22:437–442. doi: 10.1681/ASN.2010070760 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Buckley LF, Canada JM, Carbone S, Trankle CR, Kadariya D, Billingsley H, Wohlford GF, Kirkman DL, Abbate A, Van Tassell BW. Potential role for interleukin-1 in the cardio-renal syndrome. Eur J Heart Fail 2019;21:385–386. doi: 10.1002/ejhf.1403 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nowak KL, Chonchol M, Ikizler TA, Farmer-Bailey H, Salas N, Chaudhry R, Wang W, Smits G, Tengesdal I, Dinarello CA, et al. IL-1 Inhibition and Vascular Function in CKD. J Am Soc Nephrol 2017;28:971–980. doi: 10.1681/ASN.2016040453 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hung AM, Tsuchida Y, Nowak KL, Sarkar S, Chonchol M, Whitfield V, Salas N, Dikalova A, Yancey PG, Huang J, et al. IL-1 Inhibition and Function of the HDL-Containing Fraction of Plasma in Patients with Stages 3 to 5 CKD. Clinical Journal of the American Society of Nephrology 2019;14:702–711. doi: 10.2215/cjn.04360418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ridker PM, MacFadyen JG, Glynn RJ, Koenig W, Libby P, Everett BM, Lefkowitz M, Thuren T, Cornel JH. Inhibition of Interleukin-1beta by Canakinumab and Cardiovascular Outcomes in Patients With Chronic Kidney Disease. J Am Coll Cardiol 2018;71:2405–2414. doi: 10.1016/j.jacc.2018.03.490 [DOI] [PubMed] [Google Scholar]
- 60.Hong DS, Janku F, Naing A, Falchook GS, Piha-Paul S, Wheler JJ, Fu S, Tsimberidou AM, Stecher M, Mohanty P, et al. Xilonix, a novel true human antibody targeting the inflammatory cytokine interleukin-1 alpha, in non-small cell lung cancer. Invest New Drugs 2015;33:621–631. doi: 10.1007/s10637-015-0226-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.El Sayed H, Kerensky R, Stecher M, Mohanty P, Davies M. A randomized phase II study of Xilonix, a targeted therapy against interleukin 1α, for the prevention of superficial femoral artery restenosis after percutaneous revascularization. J Vasc Surg 2016;63:133–141.e131. doi: 10.1016/j.jvs.2015.08.069 [DOI] [PubMed] [Google Scholar]
- 62.Ridker PM, Devalaraja M, Baeres FMM, Engelmann MDM, Hovingh GK, Ivkovic M, Lo L, Kling D, Pergola P, Raj D, et al. IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 2021;397:2060–2069. doi: 10.1016/s0140-6736(21)00520-1 [DOI] [PubMed] [Google Scholar]
- 63.Ridker PM. From RESCUE to ZEUS: will interleukin-6 inhibition with ziltivekimab prove effective for cardiovascular event reduction? Cardiovasc Res 2021;117:e138–e140. doi: 10.1093/cvr/cvab231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Swerdlow DI, Holmes MV, Kuchenbaecker KB, Engmann JE, Shah T, Sofat R, Guo Y, Chung C, Peasey A, Pfister R, et al. The interleukin-6 receptor as a target for prevention of coronary heart disease: a mendelian randomisation analysis. Lancet 2012;379:1214–1224. doi: 10.1016/s0140-6736(12)60110-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Cupido AJ, Asselbergs FW, Natarajan P, Ridker PM, Hovingh GK, Schmidt AF. Dissecting the IL-6 pathway in cardiometabolic disease: A Mendelian randomization study on both IL6 and IL6R. Br J Clin Pharmacol 2022;88:2875–2884. doi: 10.1111/bcp.15191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Yu Z, Zekavat SM, Honigberg MC, Natarajan P. Genetic IL-6 Signaling Modifies Incident Coronary Artery Disease Risk in Chronic Kidney Disease. J Am Coll Cardiol 2022;79:415–416. doi: 10.1016/j.jacc.2021.11.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Broch K, Anstensrud AK, Woxholt S, Sharma K, Tøllefsen IM, Bendz B, Aakhus S, Ueland T, Amundsen BH, Damås JK, et al. Randomized Trial of Interleukin-6 Receptor Inhibition in Patients With Acute ST-Segment Elevation Myocardial Infarction. J Am Coll Cardiol 2021;77:1845–1855. doi: 10.1016/j.jacc.2021.02.049 [DOI] [PubMed] [Google Scholar]
- 68.Kleveland O, Kunszt G, Bratlie M, Ueland T, Broch K, Holte E, Michelsen AE, Bendz B, Amundsen BH, Espevik T, et al. Effect of a single dose of the interleukin-6 receptor antagonist tocilizumab on inflammation and troponin T release in patients with non-ST-elevation myocardial infarction: a double-blind, randomized, placebo-controlled phase 2 trial. Eur Heart J 2016;37:2406–2413. doi: 10.1093/eurheartj/ehw171 [DOI] [PubMed] [Google Scholar]
- 69.Robertson J, Peters MJ, McInnes IB, Sattar N. Changes in lipid levels with inflammation and therapy in RA: a maturing paradigm. Nature Reviews Rheumatology 2013;9:513–523. doi: 10.1038/nrrheum.2013.91 [DOI] [PubMed] [Google Scholar]
- 70.Pierini FS, Botta E, Soriano ER, Martin M, Boero L, Meroño T, Saez MS, Lozano Chiappe E, Cerda O, Citera G, et al. Effect of Tocilizumab on LDL and HDL Characteristics in Patients with Rheumatoid Arthritis. An Observational Study. Rheumatol Ther 2021;8:803–815. doi: 10.1007/s40744-021-00304-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ruiz S, Pergola PE, Zager RA, Vaziri ND. Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease. Kidney Int 2013;83:1029–1041. doi: 10.1038/ki.2012.439 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Pergola PE, Raskin P, Toto RD, Meyer CJ, Huff JW, Grossman EB, Krauth M, Ruiz S, Audhya P, Christ-Schmidt H, et al. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N Engl J Med 2011;365:327–336. doi: 10.1056/NEJMoa1105351 [DOI] [PubMed] [Google Scholar]
- 73.de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, Christ-Schmidt H, Goldsberry A, Houser M, Krauth M, Lambers Heerspink HJ, et al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 2013;369:2492–2503. doi: 10.1056/NEJMoa1306033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Engelen SE, Robinson AJB, Zurke Y-X, Monaco C. Therapeutic strategies targeting inflammation and immunity in atherosclerosis: how to proceed? Nat Rev Cardiol 2022;19:522–542. doi: 10.1038/s41569-021-00668-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sehgal SN. Sirolimus: its discovery, biological properties, and mechanism of action. Transplant Proc 2003;35:7s–14s. doi: 10.1016/s0041-1345(03)00211-2 [DOI] [PubMed] [Google Scholar]
- 76.Silva AL, Fusco DR, Nga HS, Takase HM, Bravin AM, Contti MM, Valiatti MF, de Andrade LGM. Effect of sirolimus on carotid atherosclerosis in kidney transplant recipients: data derived from a prospective randomized controlled trial. Clin Kidney J 2018;11:846–852. doi: 10.1093/ckj/sfy041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Murakami N, Riella LV, Funakoshi T. Risk of Metabolic Complications in Kidney Transplantation After Conversion to mTOR Inhibitor: A Systematic Review and Meta-Analysis. American Journal of Transplantation 2014;14:2317–2327. doi: 10.1111/ajt.12852 [DOI] [PubMed] [Google Scholar]
- 78.Cronstein BN, Aune TM. Methotrexate and its mechanisms of action in inflammatory arthritis. Nat Rev Rheumatol 2020;16:145–154. doi: 10.1038/s41584-020-0373-9 [DOI] [PubMed] [Google Scholar]
- 79.Ridker PM, Everett BM, Pradhan A, MacFadyen JG, Solomon DH, Zaharris E, Mam V, Hasan A, Rosenberg Y, Iturriaga E, et al. Low-Dose Methotrexate for the Prevention of Atherosclerotic Events. New England Journal of Medicine 2018;380:752–762. doi: 10.1056/NEJMoa1809798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sparks JA, Vanni KMM, Sparks MA, Xu C, Santacroce LM, Glynn RJ, Ridker PM, Solomon DH. Effect of Low-Dose Methotrexate on eGFR and Kidney Adverse Events: A Randomized Clinical Trial. J Am Soc Nephrol 2021;32:3197–3207. doi: 10.1681/asn.2021050598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hayashi K, Sada K-E, Asano Y, Asano SH, Yamamura Y, Ohashi K, Morishita M, Watanabe H, Narazaki M, Matsumoto Y, et al. Risk of higher dose methotrexate for renal impairment in patients with rheumatoid arthritis. Scientific Reports 2020;10:18715. doi: 10.1038/s41598-020-75655-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Nidorf SM, Fiolet ATL, Mosterd A, Eikelboom JW, Schut A, Opstal TSJ, The SHK, Xu X-F, Ireland MA, Lenderink T, et al. Colchicine in Patients with Chronic Coronary Disease. New England Journal of Medicine 2020;383:1838–1847. doi: 10.1056/NEJMoa2021372 [DOI] [PubMed] [Google Scholar]
- 83.Tardif J-C, Kouz S, Waters DD, Bertrand OF, Diaz R, Maggioni AP, Pinto FJ, Ibrahim R, Gamra H, Kiwan GS, et al. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. New England Journal of Medicine 2019;381:2497–2505. doi: 10.1056/NEJMoa1912388 [DOI] [PubMed] [Google Scholar]
- 84.LODOCO (colchicine) [package insert] Parsippany, NJ: AGEPHA Pharma USA, LLC; 2023. In. [Google Scholar]
- 85.Nelson K, Fuster V, Ridker PM. Low-Dose Colchicine for Secondary Prevention of Coronary Artery Disease. Journal of the American College of Cardiology 2023;82:648–660. doi: doi: 10.1016/j.jacc.2023.05.055 [DOI] [PubMed] [Google Scholar]
- 86.Virani SS, Newby LK, Arnold SV, Bittner V, Brewer LC, Demeter SH, Dixon DL, Fearon WF, Hess B, Johnson HM, et al. 2023. AHA/ACC/ACCP/ASPC/NLA/PCNA Guideline for the Management of Patients With Chronic Coronary Disease: A Report of the American Heart Association/American College of Cardiology Joint Committee on Clinical Practice Guidelines. Circulation 0. doi: doi: 10.1161/CIR.0000000000001168 [DOI] [PubMed] [Google Scholar]
- 87.Visseren FLJ, Mach F, Smulders YM, Carballo D, Koskinas KC, Bäck M, Benetos A, Biffi A, Boavida J-M, Capodanno D, et al. 2021 ESC Guidelines on cardiovascular disease prevention in clinical practice: Developed by the Task Force for cardiovascular disease prevention in clinical practice with representatives of the European Society of Cardiology and 12 medical societies With the special contribution of the European Association of Preventive Cardiology (EAPC). European Heart Journal 2021;42:3227–3337. doi: 10.1093/eurheartj/ehab484 [DOI] [PubMed] [Google Scholar]
- 88.Deftereos SG, Beerkens FJ, Shah B, Giannopoulos G, Vrachatis DA, Giotaki SG, Siasos G, Nicolas J, Arnott C, Patel S, et al. Colchicine in Cardiovascular Disease: In-Depth Review. Circulation 2022;145:61–78. doi: doi: 10.1161/CIRCULATIONAHA.121.056171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Silvis MJM, Fiolet ATL, Opstal TSJ, Dekker M, Suquilanda D, Zivkovic M, Duyvendak M, The SHK, Timmers L, Bax WA, et al. Colchicine reduces extracellular vesicle NLRP3 inflammasome protein levels in chronic coronary disease: A LoDoCo2 biomarker substudy. Atherosclerosis 2021;334:93–100. doi: 10.1016/j.atherosclerosis.2021.08.005 [DOI] [PubMed] [Google Scholar]
- 90.Yaxley J, Yaxley W, Scott T. Safety and adverse effect profile of colchicine in renal impairment: a systematic review of randomised trials. J Renal Inj Prev 2020;9:e28–e28. doi: 10.34172/jrip.2020.28 [DOI] [Google Scholar]
- 91.Wang Y, Peng X, Hu J, Luo T, Wang Z, Cheng Q, Mei M, He W, Peng C, Ma L, et al. Low-dose colchicine in type 2 diabetes with microalbuminuria: A double-blind randomized clinical trial. J Diabetes 2021;13:827–836. doi: 10.1111/1753-0407.13174 [DOI] [PubMed] [Google Scholar]
- 92.Kim HW, Joo YS, Yun HR, Kim JY, Jhee JH, Roh YH, Park JT, Chang TI, Yoo TH, Kang SW, et al. Colchicine use and the risk of CKD progression: a multicentre nested case-control study. Rheumatology (Oxford) 2022;61:4314–4323. doi: 10.1093/rheumatology/keac077 [DOI] [PubMed] [Google Scholar]
- 93.Goicoechea M, de Vinuesa SG, Verdalles U, Ruiz-Caro C, Ampuero J, Rincón A, Arroyo D, Luño J. Effect of allopurinol in chronic kidney disease progression and cardiovascular risk. Clin J Am Soc Nephrol 2010;5:1388–1393. doi: 10.2215/cjn.01580210 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Badve SV, Pascoe EM, Tiku A, Boudville N, Brown FG, Cass A, Clarke P, Dalbeth N, Day RO, de Zoysa JR, et al. Effects of Allopurinol on the Progression of Chronic Kidney Disease. N Engl J Med 2020;382:2504–2513. doi: 10.1056/NEJMoa1915833 [DOI] [PubMed] [Google Scholar]
- 95.Doria A, Galecki AT, Spino C, Pop-Busui R, Cherney DZ, Lingvay I, Parsa A, Rossing P, Sigal RJ, Afkarian M, et al. Serum Urate Lowering with Allopurinol and Kidney Function in Type 1 Diabetes. N Engl J Med 2020;382:2493–2503. doi: 10.1056/NEJMoa1916624 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kimura K, Hosoya T, Uchida S, Inaba M, Makino H, Maruyama S, Ito S, Yamamoto T, Tomino Y, Ohno I, et al. Febuxostat Therapy for Patients With Stage 3 CKD and Asymptomatic Hyperuricemia: A Randomized Trial. Am J Kidney Dis 2018;72:798–810. doi: 10.1053/j.ajkd.2018.06.028 [DOI] [PubMed] [Google Scholar]
- 97.Kojima S, Matsui K, Hiramitsu S, Hisatome I, Waki M, Uchiyama K, Yokota N, Tokutake E, Wakasa Y, Jinnouchi H, et al. Febuxostat for Cerebral and CaRdiorenovascular Events PrEvEntion StuDy. Eur Heart J 2019;40:1778–1786. doi: 10.1093/eurheartj/ehz119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kojima S, Uchiyama K, Yokota N, Tokutake E, Wakasa Y, Hiramitsu S, Waki M, Jinnouchi H, Kakuda H, Hayashi T, et al. C-reactive Protein Levels and Cardiovascular Outcomes After Febuxostat Treatment in Patients with Asymptomatic Hyperuricemia: Post-hoc Analysis of a Randomized Controlled Study. Cardiovasc Drugs Ther 2022. doi: 10.1007/s10557-022-07347-7 [DOI] [PubMed]
- 99.Major RW, Cheung CK, Gray LJ, Brunskill NJ. Statins and Cardiovascular Primary Prevention in CKD: A Meta-Analysis. Clin J Am Soc Nephrol 2015;10:732–739. doi: 10.2215/cjn.07460714 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wang J, Chen Z, Qiu Y, Wu L, Wang H, Wu L, Zhao L, Xie D. Statins Have an Anti-Inflammation in CKD Patients: A Meta-Analysis of Randomized Trials. Biomed Res Int 2022;2022:4842699. doi: 10.1155/2022/4842699 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Krane V, Wanner C. Statins, inflammation and kidney disease. Nat Rev Nephrol 2011;7:385–397. doi: 10.1038/nrneph.2011.62 [DOI] [PubMed] [Google Scholar]
- 102.Ridker PM, Danielson E, Fonseca FAH, Genest J, Gotto AM, Kastelein JJP, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG, et al. Rosuvastatin to Prevent Vascular Events in Men and Women with Elevated C-Reactive Protein. New England Journal of Medicine 2008;359:2195–2207. doi: 10.1056/NEJMoa0807646 [DOI] [PubMed] [Google Scholar]
- 103.Ridker PM, MacFadyen J, Cressman M, Glynn RJ. Efficacy of rosuvastatin among men and women with moderate chronic kidney disease and elevated high-sensitivity C-reactive protein: a secondary analysis from the JUPITER (Justification for the Use of Statins in Prevention-an Intervention Trial Evaluating Rosuvastatin) trial. J Am Coll Cardiol 2010;55:1266–1273. doi: 10.1016/j.jacc.2010.01.020 [DOI] [PubMed] [Google Scholar]
- 104.Baigent C, Landray MJ, Reith C, Emberson J, Wheeler DC, Tomson C, Wanner C, Krane V, Cass A, Craig J, et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet 2011;377:2181–2192. doi: 10.1016/s0140-6736(11)60739-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Fellström BC, Jardine AG, Schmieder RE, Holdaas H, Bannister K, Beutler J, Chae DW, Chevaile A, Cobbe SM, Grönhagen-Riska C, et al. Rosuvastatin and cardiovascular events in patients undergoing hemodialysis. N Engl J Med 2009;360:1395–1407. doi: 10.1056/NEJMoa0810177 [DOI] [PubMed] [Google Scholar]
- 106.Wanner C, Krane V, März W, Olschewski M, Mann JF, Ruf G, Ritz E. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med 2005;353:238–248. doi: 10.1056/NEJMoa043545 [DOI] [PubMed] [Google Scholar]
- 107.Krane V, Winkler K, Drechsler C, Lilienthal J, März W, Wanner C. Effect of atorvastatin on inflammation and outcome in patients with type 2 diabetes mellitus on hemodialysis. Kidney Int 2008;74:1461–1467. doi: 10.1038/ki.2008.484 [DOI] [PubMed] [Google Scholar]
- 108.Barrera-Chimal J, Girerd S, Jaisser F. Mineralocorticoid receptor antagonists and kidney diseases: pathophysiological basis. Kidney Int 2019;96:302–319. doi: 10.1016/j.kint.2019.02.030 [DOI] [PubMed] [Google Scholar]
- 109.Bakris GL, Agarwal R, Anker SD, Pitt B, Ruilope LM, Rossing P, Kolkhof P, Nowack C, Schloemer P, Joseph A, et al. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N Engl J Med 2020;383:2219–2229. doi: 10.1056/NEJMoa2025845 [DOI] [PubMed] [Google Scholar]
- 110.Pitt B, Filippatos G, Agarwal R, Anker SD, Bakris GL, Rossing P, Joseph A, Kolkhof P, Nowack C, Schloemer P, et al. Cardiovascular Events with Finerenone in Kidney Disease and Type 2 Diabetes. N Engl J Med 2021;385:2252–2263. doi: 10.1056/NEJMoa2110956 [DOI] [PubMed] [Google Scholar]
- 111.Cannavo A, Marzano F, Elia A, Liccardo D, Bencivenga L, Gambino G, Perna C, Rapacciuolo A, Cittadini A, Ferrara N, et al. Aldosterone Jeopardizes Myocardial Insulin and β-Adrenergic Receptor Signaling via G Protein-Coupled Receptor Kinase 2. Front Pharmacol 2019;10:888. doi: 10.3389/fphar.2019.00888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Bauersachs J, Jaisser F, Toto R. Mineralocorticoid Receptor Activation and Mineralocorticoid Receptor Antagonist Treatment in Cardiac and Renal Diseases. Hypertension 2015;65:257–263. doi: doi: 10.1161/HYPERTENSIONAHA.114.04488 [DOI] [PubMed] [Google Scholar]
- 113.Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, Greene T, Hou F-F, Mann JFE, McMurray JJV, Lindberg M, Rossing P, et al. Dapagliflozin in Patients with Chronic Kidney Disease. New England Journal of Medicine 2020;383:1436–1446. doi: 10.1056/NEJMoa2024816 [DOI] [PubMed] [Google Scholar]
- 114.Kim SR, Lee SG, Kim SH, Kim JH, Choi E, Cho W, Rim JH, Hwang I, Lee CJ, Lee M, et al. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat Commun 2020;11:2127. doi: 10.1038/s41467-020-15983-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Heerspink HJL, Perco P, Mulder S, Leierer J, Hansen MK, Heinzel A, Mayer G. Canagliflozin reduces inflammation and fibrosis biomarkers: a potential mechanism of action for beneficial effects of SGLT2 inhibitors in diabetic kidney disease. Diabetologia 2019;62:1154–1166. doi: 10.1007/s00125-019-4859-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Shaman AM, Bain SC, Bakris GL, Buse JB, Idorn T, Mahaffey KW, Mann JFE, Nauck MA, Rasmussen S, Rossing P, et al. Effect of the Glucagon-Like Peptide-1 Receptor Agonists Semaglutide and Liraglutide on Kidney Outcomes in Patients With Type 2 Diabetes: Pooled Analysis of SUSTAIN 6 and LEADER. Circulation 2022;145:575–585. doi: doi: 10.1161/CIRCULATIONAHA.121.055459 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Furtado RHM, Bonaca MP, Mosenzon O, Kato ET, Cahn A, et al. Comparison of the Effects of Glucagon-Like Peptide Receptor Agonists and Sodium-Glucose Cotransporter 2 Inhibitors for Prevention of Major Adverse Cardiovascular and Renal Outcomes in Type 2 Diabetes Mellitus. Circulation 2019;139:2022–2031. doi: doi: 10.1161/CIRCULATIONAHA.118.038868 [DOI] [PubMed] [Google Scholar]
- 118.Yu JH, Park SY, Lee DY, Kim NH, Seo JA. GLP-1 receptor agonists in diabetic kidney disease: current evidence and future directions. Kidney Res Clin Pract 2022;41:136–149. doi: 10.23876/j.krcp.22.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Mazidi M, Karimi E, Rezaie P, Ferns GA. Treatment with GLP1 receptor agonists reduce serum CRP concentrations in patients with type 2 diabetes mellitus: A systematic review and meta-analysis of randomized controlled trials. Journal of Diabetes and its Complications 2017;31:1237–1242. doi: 10.1016/j.jdiacomp.2016.05.022 [DOI] [PubMed] [Google Scholar]
- 120.Cottone S, Palermo A, Vaccaro F, Mulè G, Guarneri M, Arsena R, Vadalà A, Cerasola G. Inflammation and endothelial activation are linked to renal function in long-term kidney transplantation. Transpl Int 2007;20:82–87. doi: 10.1111/j.1432-2277.2006.00405.x [DOI] [PubMed] [Google Scholar]
- 121.Meier-Kriesche HU, Schold JD, Srinivas TR, Reed A, Kaplan B. Kidney transplantation halts cardiovascular disease progression in patients with end-stage renal disease. Am J Transplant 2004;4:1662–1668. doi: 10.1111/j.1600-6143.2004.00573.x [DOI] [PubMed] [Google Scholar]
- 122.Kasiske BL, Maclean JR, Snyder JJ. Acute myocardial infarction and kidney transplantation. J Am Soc Nephrol 2006;17:900–907. doi: 10.1681/ASN.2005090984 [DOI] [PubMed] [Google Scholar]
- 123.Molnar MZ, Nagy K, Remport A, Tapolyai MB, Fülöp T, Kamal F, Kovesdy CP, Mucsi I, Mathe Z. Inflammatory Markers and Outcomes in Kidney Transplant Recipients. Transplantation 2017;101:2152–2164. doi: 10.1097/tp.0000000000001548 [DOI] [PubMed] [Google Scholar]
- 124.Abedini S, Holme I, März W, Weihrauch G, Fellström B, Jardine A, Cole E, Maes B, Neumayer HH, Grønhagen-Riska C, et al. Inflammation in renal transplantation. Clin J Am Soc Nephrol 2009;4:1246–1254. doi: 10.2215/cjn.00930209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Holdaas H, Fellström B, Jardine AG, Holme I, Nyberg G, Fauchald P, Grönhagen-Riska C, Madsen S, Neumayer HH, Cole E, et al. Effect of fluvastatin on cardiac outcomes in renal transplant recipients: a multicentre, randomised, placebo-controlled trial. Lancet 2003;361:2024–2031. doi: 10.1016/s0140-6736(03)13638-0 [DOI] [PubMed] [Google Scholar]
- 126.Miller CL, Madsen JC. IL-6 Directed Therapy in Transplantation. Curr Transplant Rep 2021;8:191–204. doi: 10.1007/s40472-021-00331-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, Kakehi T. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti–IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood 2008;112:3959–3964. doi: 10.1182/blood-2008-05-155846 [DOI] [PubMed] [Google Scholar]
- 128.de Fijter JW, Holdaas H, Øyen O, Sanders JS, Sundar S, Bemelman FJ, Sommerer C, Pascual J, Avihingsanon Y, Pongskul C, et al. Early Conversion From Calcineurin Inhibitor- to Everolimus-Based Therapy Following Kidney Transplantation: Results of the Randomized ELEVATE Trial. Am J Transplant 2017;17:1853–1867. doi: 10.1111/ajt.14186 [DOI] [PubMed] [Google Scholar]
- 129.Martin-Moreno PL, Tripathi S, Chandraker A. Regulatory T Cells and Kidney Transplantation. Clin J Am Soc Nephrol 2018;13:1760–1764. doi: 10.2215/cjn.01750218 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.O’Donoghue ML, Rosenson RS, Gencer B, López JAG, Lepor NE, Baum SJ, Stout E, Gaudet D, Knusel B, Kuder JF, et al. Small Interfering RNA to Reduce Lipoprotein(a) in Cardiovascular Disease. N Engl J Med 2022;387:1855–1864. doi: 10.1056/NEJMoa2211023 [DOI] [PubMed] [Google Scholar]
- 131.Dzobo KE, Kraaijenhof JM, Stroes ESG, Nurmohamed NS, Kroon J. Lipoprotein(a): An underestimated inflammatory mastermind. Atherosclerosis 2022;349:101–109. doi: 10.1016/j.atherosclerosis.2022.04.004 [DOI] [PubMed] [Google Scholar]
- 132.Yi T, Wang J, Zhu K, Tang Y, Huang S, Shui X, Ding Y, Chen C, Lei W. Aryl Hydrocarbon Receptor: A New Player of Pathogenesis and Therapy in Cardiovascular Diseases. Biomed Res Int 2018;2018:6058784. doi: 10.1155/2018/6058784 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Vogel CF, Sciullo E, Matsumura F. Activation of inflammatory mediators and potential role of ah-receptor ligands in foam cell formation. Cardiovasc Toxicol 2004;4:363–373. doi: 10.1385/ct:4:4:363 [DOI] [PubMed] [Google Scholar]
- 134.Tumur Z, Shimizu H, Enomoto A, Miyazaki H, Niwa T. Indoxyl sulfate upregulates expression of ICAM-1 and MCP-1 by oxidative stress-induced NF-kappaB activation. Am J Nephrol 2010;31:435–441. doi: 10.1159/000299798 [DOI] [PubMed] [Google Scholar]
- 135.Safe S, Cheng Y, Jin UH. The Aryl Hydrocarbon Receptor (AhR) as a Drug Target for Cancer Chemotherapy. Curr Opin Toxicol 2017;2:24–29. doi: 10.1016/j.cotox.2017.01.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Tarkin JM, Joshi FR, Rudd JHF. PET imaging of inflammation in atherosclerosis. Nat Rev Cardiol 2014;11:443–457. doi: 10.1038/nrcardio.2014.80 [DOI] [PubMed] [Google Scholar]
- 137.Toner YC, Ghotbi AA, Naidu S, Sakurai K, van Leent MMT, Jordan S, Ordikhani F, Amadori L, Sofias AM, Fisher EL, et al. Systematically evaluating DOTATATE and FDG as PET immuno-imaging tracers of cardiovascular inflammation. Scientific Reports 2022;12:6185. doi: 10.1038/s41598-022-09590-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Bajaj NS, Singh A, Zhou W, Gupta A, Fujikura K, Byrne C, Harms HJ, Osborne MT, Bravo P, Andrikopolou E, et al. Coronary Microvascular Dysfunction, Left Ventricular Remodeling, and Clinical Outcomes in Patients With Chronic Kidney Impairment. Circulation 2020;141:21–33. doi: 10.1161/CIRCULATIONAHA.119.043916 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Oikonomou EK, Marwan M, Desai MY, Mancio J, Alashi A, Hutt Centeno E, Thomas S, Herdman L, Kotanidis CP, Thomas KE, et al. Non-invasive detection of coronary inflammation using computed tomography and prediction of residual cardiovascular risk (the CRISP CT study): a post-hoc analysis of prospective outcome data. Lancet 2018;392:929–939. doi: 10.1016/s0140-6736(18)31114-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
