ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Inflammation in patients with kidney function impairment

Inflammation in patients with kidney function impairment
Literature review current through: Jan 2024.
This topic last updated: Aug 29, 2023.

INTRODUCTION — Acute and chronic inflammatory processes are common in individuals with chronic kidney disease (CKD) and especially end-stage kidney disease (ESKD). This is due to many underlying factors, including the uremic milieu, elevated levels of circulating proinflammatory cytokines, oxidative stress, carbonyl stress, protein-energy wasting (PEW), enhanced incidence of infections (especially dialysis access related), and others. Although the definition of inflammation is unclear in this setting, CKD-associated chronic inflammation, as indicated by increased C-reactive protein (CRP) levels above 5 mg/L over at least three months, has been reported in 30 to 60 percent of North American and European dialysis patients, with dialysis patients in Asian countries possibly having a lower prevalence [1-7].

The acute-phase response is a major pathophysiologic phenomenon that accompanies inflammation. With this reaction, normal homeostatic mechanisms are replaced by new set points that presumably contribute to defensive or adaptive capabilities.

Acute-phase proteins are defined as those proteins whose plasma concentrations increase (positive acute-phase proteins), such as CRP, or decrease (negative acute-phase proteins), such as albumin, during inflammatory states. Measurement of the levels of these proteins is frequently utilized to define the presence and/or degree of inflammation in a given patient. A number of inflammatory markers have been studied in patients with CKD (table 1). (See "Acute phase reactants".)

Despite its name, the "acute"-phase response can persist over months to years and become chronic. In such states of chronic inflammation, positive acute-phase proteins including CRP (normal range <1 mg/L) may be slightly but persistently increased, which can predispose to an increased risk of atherosclerotic cardiovascular disease (CVD; CRP 1 to 3 mg/L) [8]. However, in many patients with CKD, especially in maintenance dialysis patients, serum CRP levels are persistently between 5 and 50 mg/L, although they may fluctuate widely [8]. During acute and fulminant infections, such as acute osteomyelitis, serum CRP is not uncommonly above 50 mg/L (table 2).

Among patients with CKD, the presence of an inflammatory state may also be closely related to accelerated atherogenesis, protein-energy wasting (PEW), and anemia [9-11]:

Kidney function impairment alone is reported to be a risk factor for atherosclerosis and CVD. This may be due in part to the frequent concurrence of inflammation. (See "Chronic kidney disease and coronary heart disease".)

Increasing attention has focused upon inflammation as a possible cause of the wasting syndrome in ESKD as well as anemia refractory to therapy with erythropoiesis-stimulating agents (ESAs).

The pathogenesis and consequences of inflammatory processes among patients with kidney dysfunction will be discussed in this topic review.

INFLAMMATION AND KIDNEY DISEASE — A generalized increase in the inflammatory response in patients with decreased kidney function may occur via the following mechanisms:

Decreased renal clearance of proinflammatory cytokines

Volume overload with endotoxemia

Oxidative and carbonyl stress

Decreased levels of antioxidants

Increased presence of comorbid conditions

Decreased clearance of proinflammatory cytokines — The kidneys are responsible for clearing proinflammatory cytokines and bacterial antigens from circulation [12]. Deteriorating kidney function may enhance overall inflammatory responses because of the decreased renal clearance of factors that are directly or indirectly involved in inflammation. As an example, the serum half-lives of proinflammatory cytokines, tumor necrosis factor alpha (TNF-alpha), and interleukin (IL) 1 are greater in animals without kidney function than those with kidney function [13,14].

In humans, declining kidney function may also affect the levels of additional inflammatory molecules as serum C-reactive protein (CRP), IL-6, and hyaluronan levels are inversely correlated with creatinine clearance [15,16], although inflammation may have a bidirectional relationship with kidney function, being not only a consequence but also a cause for loss of glomerular filtration rate (GFR) [17,18]. In addition, among patients with end-stage kidney disease (ESKD) who have residual kidney function, higher serum CRP concentrations are observed among those with relatively less native kidney function [19,20]. Increased production of cytokines may also be observed. Furthermore, the loss of function in various tissues due to enhanced proinflammatory cytokines can in turn result in release of endogenous damage-associated molecular patterns (DAMPs), such as mitochondrial DNA, which can further drive systemic inflammation in ESKD [21]. (See 'Oxidative and carbonyl stress' below and "Uremic toxins".)

Volume overload with endotoxemia — Vascular congestion due to fluid overload in patients with kidney function impairment may result in altered permeability of the gastrointestinal tract, thereby allowing the translocation of gut-derived toxic products such as endotoxins and bacterial DNA fragments into the systemic circulation [22]. These processes may in turn stimulate immune cells and the increased release of proinflammatory cytokines [23-26].

Oxidative and carbonyl stress — Increased production of cytokines due to oxidative stress is also observed among patients with kidney failure [27,28]. Oxidative stress, which occurs when there is excessive free-radical production or low antioxidant levels, could be an important condition for the development of endothelial dysfunction, inflammation, and atherogenesis [28-30] (see "Coronary endothelial dysfunction: Clinical aspects"). Lower plasmalogen levels, an indicator of such stress, have been reported in malnourished and inflamed patients with chronic kidney disease (CKD) [31].

With kidney failure, molecules that are not cytokines may also accumulate and provoke an inflammatory response. As an example, advanced glycosylated end products (AGEs), which result from carbonyl stress, can clearly initiate inflammation in patients with kidney failure [32,33]. Other uremic toxins, such as indoxyl sulphate, can also evoke inflammatory pathways either directly or through reactive oxygen species (ROS)-mediated activation of nuclear factor kappa B (NF-kB) signaling [28,34]. (See "Uremic toxins".)

Decreased antioxidants — The oral intake or the circulating levels of some antioxidants are lower than normal in patients with CKD and ESKD [35]. An acute-phase response is also associated with decreased plasma levels of several antioxidants, such as serum vitamin C concentrations [27,36]. Low serum vitamin C levels are in turn associated with increased cardiovascular morbidity and mortality [37]. Nuclear factor erythroid 2–related factor 2 (Nrf2) is a transcription factor that plays a key role in adaptive stress response mechanisms against oxidative stress and inflammation [38-42]. The reduced antioxidant activity of Nrf2 coupled with an upregulation of NF-kB is typically seen in the uremic milieu, which can further drive inflammation [43,44].

Comorbid conditions — The frequent occurrence of comorbid illnesses in patients with kidney disease enhances the hypercatabolic state and the development of inflammation [45]. An example is periodontal disease [46]. Even in the absence of overt clinical illness, an inflammatory process may be ongoing and associated with an acute-phase response in this setting [47]. In addition, an increased susceptibility to infections has been described in patients with CKD and ESKD, which may be partly due to impaired immunity, uremia, old age, and comorbid conditions [48-50].

Changes in gut microbiota and diet — Changes in diet composition and in gut microbiota have also been linked to alterations in intestinal permeability and production of uremic toxins, leading to inflammation [51]. This raises the possibility that gastrointestinal alterations, such as constipation or pathologic alterations in the composition of intestinal milieu, may have an effect on kidney function and on cardiovascular outcomes [52-54]. Changes in gut microbiota composition may also be related to protein-energy wasting (PEW) in patients with ESKD [55].

Other factors — A systemic autoimmune condition (which may have caused the kidney disease), clonal hematopoiesis, epigenetic (eg, microRNA and DNA methylation) and genetic factors, premature cellular aging, an unrecognized persistent infection, periodontal diseases, and atherosclerosis may also underlie inflammation among patients with decreased kidney function or ESKD [56-65].

End-stage kidney disease

Hemodialysis — In addition to the previously mentioned causes that may underlie and/or enhance ongoing inflammation in patients with CKD, the following conditions may chronically aggravate inflammatory processes in those undergoing maintenance hemodialysis:

Exposure to dialysis tubing and dialysis membranes, particularly less biocompatible membranes (eg, cuprophane membranes) [66,67]. (See "Clinical consequences of hemodialysis membrane biocompatibility".)

Poor quality of dialysis water and back-filtration or back-diffusion of contaminants, resulting in possible exposure to endotoxins. (See "Contaminants in water used for hemodialysis".)

The presence of foreign bodies (such as polytetrafluoroethylene [PTFE] chronic access grafts) or an intravenous catheter, which may harbor chronic or recurrent latent infection via the formation of a biofilm [68-73].

A clotted arteriovenous (AV) graft that is not excised.

Peritoneal dialysis — Factors unique to patients on peritoneal dialysis may also enhance chronic inflammation [74,75]:

Episodes of overt or latent peritonitis or peritoneal dialysis catheter-related infections [6,74,76].

Constant exposure to peritoneal dialysis solution, which may include bioincompatible substances or endotoxins [74,76].

Loss of residual kidney function and fluid overload [74]. However, inflammation could be also be a cause of the loss of residual kidney function [77].

Presence of a failed kidney transplant — A chronic inflammatory state has been noted in patients who return to dialysis after a failed kidney transplant but in whom a transplant nephrectomy has not been performed [78,79]. Symptoms and signs of inflammation may abate with kidney removal. (See "Kidney transplantation in adults: Management of the patient with a failed kidney transplant".)

GENERAL MARKERS OF INFLAMMATION — There is no consensus approach to assess the degree of severity of inflammation in individuals with kidney disease (see "Acute phase reactants"). Routine monitoring of inflammatory markers in all patients with end-stage kidney disease (ESKD) or chronic kidney disease (CKD) is not recommended by most guidelines, although it may improve patient management. Periodic measurement of C-reactive protein (CRP) or interleukin (IL) 6 to assess severity and frequency of inflammation is reasonable in select patients as part of an individualized work-up prompted by specific clinical characteristics, such as protein-energy wasting (PEW).

Several measures of general inflammation are used in patients with CKD or ESKD as well as in others [80-90]. These include the following proteins:

Serum levels of positive acute-phase reactants, such as serum CRP, ferritin, or fibrinogen, are commonly elevated during an acute episode of inflammation. There is increasing literature on elevated serum CRP and increased cardiovascular risk, a relationship that is independent of other coronary risk factors. (See "C-reactive protein in cardiovascular disease".)

Serum levels of negative acute-phase reactants, such as albumin or transferrin, decrease during an inflammatory process [81,82]. Many negative acute-phase reactants are also traditionally utilized as nutritional markers since their serum levels are decreased with a decline in nutritional status.

Since episodes of occult or overt inflammation may be chronic or recurrent, they have occasionally been referred to as representing a "chronic" acute-phase response [83,84]. Such chronic inflammatory responses are associated with an elevation of serum acute-phase proteins including CRP, erythrocyte sedimentation rate, serum amyloid A, and some proinflammatory cytokines including a variety of ILs. Among the proinflammatory cytokines, IL-6 may have a central role in the pathophysiology of inflammation in patients with kidney disease [85].

No consensus exists as to what threshold levels of inflammatory markers optimally distinguish noninflamed from inflamed patients with ESKD, as well as what exact markers should be measured. Based upon mean values and/or possible correlation with outcomes, several investigators have proposed different serum levels of various markers [1,80,91-96]:

In a survival study of 91 patients on hemodialysis, a serum CRP level >5 mg/L (>0.5 mg/dL, as measured by end-point nephelometry) was proposed as a sign of inflammation [1]. This was derived from the observation that the mean CRP values among patients who were alive or had died at study end were originally 3.4 and 10.1 mg/L, respectively.

In a prospective report of 240 patients on peritoneal dialysis with an initial median serum CRP level of 7 mg/L, the CRP level was significantly higher among the 84 patients who experienced a cardiovascular event during the subsequent 41 months (27 mg/L) [91].

In a third series, the mean serum IL-6 concentrations among patients on hemodialysis or peritoneal dialysis were 7.2 and 5.6 pg/mL, respectively [92]. When levels were categorized by quartiles, the group with the highest initial level had the highest mortality, a finding observed in both patients on hemodialysis and those on peritoneal dialysis.

In a review of 230 hemodialysis patients and 28 nondialytic controls, serum concentrations of IL-1, 2, 4, 5, 6, 12, and 13 as well as tumor necrosis factor alpha (TNF-alpha) were significantly increased in the dialysis group [80].

There are some patients with ESKD in whom the measurement of such inflammatory markers may help guide clinical decision making. The measurement of serum CRP and IL-6, for example, may help distinguish the relative contributions of inflammation and malnutrition to the development of hypoalbuminemia [11,97].

In addition, the serum measurement of inflammatory markers may help determine whether the presence of hyperferritinemia in patients on dialysis is due to iron overload or inflammation; this may be particularly helpful when the serum ferritin level is >800 ng/mL but the serum transferrin saturation is within the normal range (<50 percent) [98]. The association between increased mortality and serum ferritin levels >800 ng/mL appears to be primarily due to the confounding effect of inflammation [99]. (See "Diagnosis of iron deficiency in chronic kidney disease" and "Treatment of iron deficiency in patients with nondialysis chronic kidney disease (CKD)" and "Treatment of iron deficiency in patients on dialysis".)

DOES INFLAMMATION LEAD TO PROTEIN-ENERGY WASTING? — Protein-energy wasting (PEW) [100], which is present in a large proportion of patients with end-stage kidney disease (ESKD), is a strong risk factor for increased hospitalization and mortality in patients undergoing maintenance hemodialysis or peritoneal dialysis [11,83,101]. The impact of PEW on dialysis outcome may lead to a paradoxical risk factor reversal or reverse epidemiology of risk factors [102,103]. Among patients on dialysis, for example, a low (and not a high) body mass index (BMI) or a low (and not a high) serum cholesterol concentration is associated with increased mortality [11,102,104]. (See "Pathogenesis and treatment of malnutrition in patients on maintenance hemodialysis" and "Patient survival and maintenance dialysis".)

Several investigators suggest that PEW is largely a consequence of chronic inflammatory processes in patients with ESKD [9,10,84]. Although proinflammatory cytokines may be a common link between malnutrition and inflammation, additional factors such as oxidative stress, carbonyl stress, uremic toxins, and others may also play roles [29,30]. Thus, the degree to which there is a link between PEW and inflammation and the independent effect of each upon adverse outcomes in patients with kidney diseases are unclear.

The following two sections provide an overview of the data for and against a relationship between inflammation and malnutrition.

Positive observations — The hypothesis that PEW is a consequence of chronic inflammatory processes in patients with ESKD is supported by the following [9,10,84]:

Inflammation is associated with a rise in plasma and probably tissue levels of catabolic cytokines; plasma elevations of inflammatory proteins and catabolic cytokines are commonly observed in nondialyzed patients with advanced chronic kidney function impairment and in patients on dialysis. One such cytokine, tumor necrosis factor alpha (TNF-alpha), promotes catabolic processes (engendering both protein degradation and suppression of protein synthesis) and also induces anorexia [11,105].

Some patients on dialysis with chronic inflammation develop weight loss and a negative protein balance, despite an intact appetite. In this setting, there may be a shift in protein synthesis from muscle to acute-phase proteins, as well as an overall decrease in protein synthesis and increase in protein degradation, as kidney function declines. Such patients also appear to lose more body weight during dialysis when compared with those without discernible inflammatory processes [106].

Albumin synthesis is suppressed when serum C-reactive protein (CRP) is elevated [10,84]. In patients with chronic kidney disease (CKD), serum albumin decreases and proinflammatory cytokines accumulate as kidney function deteriorates [15,16,19,107,108]. Among well-dialyzed patients, activation of the acute-phase response also correlates with lower serum albumin levels due to decreased albumin synthesis [109].

Inflammation may lead to hypocholesterolemia, a strong mortality risk factor in patients on dialysis and a marker of poor nutritional status [104,110]. This was best shown in a prospective study of 823 patients who were categorized by either the presence or absence of inflammation/malnutrition, which was defined by a low serum albumin concentration or elevated levels of CRP or interleukin (IL) 6 [104]. Among all patients, as well as those with inflammation, lower cholesterol levels were associated with higher mortality. By comparison, higher cholesterol levels in those without markers of inflammation/malnutrition were associated with higher mortality.

Thus, chronic inflammation may be a principal factor that causally links PEW to increased morbidity and mortality among patients on dialysis. The term "malnutrition-inflammation-complex syndrome" (MICS) suggests the purported close association between PEW and inflammation in patients on dialysis [11,111,112]. Alternatively, "malnutrition-inflammation-atherosclerosis" (MIA) has been used to emphasize the importance of atherosclerotic diseases as a major consequence of the syndrome [113].

Several scoring systems have been proposed to assess the degree of MICS or MIA in dialysis patients, such as the "malnutrition-inflammation score" (MIS) [111]. This correlates strongly with both measures of nutritional status and inflammation and anemia. Similar associations of MIS with lower kidney function and with nutritional status have been described in patients with nondialysis-dependent CKD and kidney transplant recipients [114].

The MIS is also significantly associated with hospitalization rates and mortality in hemodialysis patients [111] since it is essentially a modified version of the Subjective Global Assessment of Nutrition (SGA) [83]; the SGA is recommended in the National Kidney Foundation Kidney Disease Outcomes Quality Initiative (KDOQI) guidelines to be used routinely to assess the nutritional status of dialysis patients. The SGA also helps measure the degree of inflammation and illness severity [83,111,115].

Negative observations — There is also evidence that does not support inflammation as the principal cause of PEW:

Serum albumin and prealbumin levels and other indicators of protein-energy nutritional status obviously correlate with protein intake and are independent of inflammatory status [116]. The serum albumin decreases, although only modestly, among normal individuals with PEW induced by reducing their nutrient intake or in malnourished hemodialysis patients fed low-protein diets, suggesting that serum albumin is a direct reflection of protein intake [108,117,118].

In dialysis patients, the association of serum albumin and CRP is hardly precise, with the reported correlation coefficients usually less than 0.5 [108,117].

Unlike serum CRP levels, serum albumin concentrations usually do not fluctuate on a month-to-month basis [84].

In some studies, the provision of adequate nutrition without management of inflammation improves hypoalbuminemia and clinical outcome [119,120].

These considerations, although not conclusive, indicate that factors other than the catabolic consequences of inflammation also affect serum albumin and other nutritional measures. Nutrient intake is obviously one such factor.

Malnutrition may lead to inflammation — There are also some data suggesting that malnutrition can lead to inflammation:

Malnourished patients on dialysis may be deficient of antioxidants such as vitamin C or carotenoids [35], which may lead to increased oxidative stress and inflammation.

PEW may decrease host resistance and predispose to infection, which is clearly an inflammatory disorder. Certain nutrients, such as arginine and glutamine, may enhance the immune response [121]. Moreover, preliminary data suggest that levocarnitine may protect against endotoxins and also suppress elaboration of TNF-alpha from monocytes [122].

Hypocholesterolemia, as a reflection of general hypolipoproteinemia in malnourished patients on dialysis, may mitigate the ability to remove circulating endotoxins. Based upon the lipoprotein-endotoxin hypothesis, there is an optimum serum lipoprotein concentration below which lipid reduction is detrimental because of the decreased ability of lipoproteins to bind lipopolysaccharide; this, in turn, may prevent lipoproteins from decreasing the detrimental effects of endotoxin [123].

Summary — PEW may be associated with a poor outcome in patients on dialysis because of inflammation. However, existing data are consistent with the possibility that nutrient intake may also independently affect outcome.

CHRONIC INFLAMMATION AND OUTCOMES — Increased levels of markers of inflammation and malnutrition predict a poor outcome in patients with end-stage kidney disease (ESKD) and chronic kidney disease (CKD). Despite this, neither infection, which usually leads to an overt inflammatory response, or malnutrition are common causes of death. Rather, most individuals with CKD die of cardiovascular diseases (CVD). Thus, the purported link from underlying inflammation to decreased survival among patients with kidney disease, if it exists, should be most strongly observed for inflammation and atherosclerosis, rather than for inflammation and infection or malnutrition.

Perhaps the best evidence supporting the importance of inflammation in the pathogenesis of atherosclerosis comes from the observation among patients without kidney disease that markers of increased or decreased systemic inflammation are directly associated with an enhanced risk of atherosclerosis.

Although the evidence is less clear, patients with ESKD with coronary heart disease and enhanced cardiovascular risk and mortality frequently have similar elevated levels of acute-phase reactants [3]:

Increased serum C-reactive protein (CRP) levels in patients with ESKD are as strongly associated with cardiovascular and all-cause mortality as decreased serum albumin levels [1,2,124,125].

Increased serum levels of interleukin (IL) 6 are associated with increased dialysis mortality [110], and progression of carotid atherosclerosis during dialysis may be related to increased IL-6 levels [126].

Increasing clinical evidence also suggests that inflammation and oxidation stress are associated with adverse outcomes among predialysis patients [127-132]. This was perhaps best shown in a prospective study of 80 nondiabetic predialysis patients in whom the effect of different conventional and nonconventional risk factors on cardiovascular events was examined [130]. At follow-up at a median period of seven years, 21 patients developed adverse outcomes due to coronary, cerebral, or peripheral artery occlusion. Upon multivariate analysis, an adverse cardiovascular outcome was independently associated with increased age and elevated CRP, fibrinogen, and advanced oxidation protein product levels. Elevated plasma levels of fibrinogen and TNF-alpha have also been linked to rapid loss of kidney function in patients with CKD [133].

One study described the role of chemokines (which are 8 to 14 kDa proteins that regulate leukocyte trafficking during inflammatory reactions) in the all-cause and cardiovascular mortality associated with elevated CRP levels in patients with ESKD [134,135]. In this Mendelian randomization study of Dutch and Swedish dialysis patients, there was a statistical interaction between the presence of a nonfunctioning allele of the chemokine receptor 5 (CCR5) and all-cause and cardiovascular mortality associated with elevated CRP level. Patients with the allele appeared unaffected by the higher cardiovascular mortality seen in association with elevated CRP, interpreted as a potential protective effect by the nonfunctioning gene. Noncardiovascular mortality appeared increased in patients with higher CRP levels irrespective of their CCR5 polymorphism, consistent with the hypothesis that CCR5 is involved in the process of atherosclerosis but also suggesting that the effects of inflammation may reach beyond the process of atherosclerosis and affect noncardiovascular mortality as well as cardiovascular mortality in ESKD [135].

In addition to atherosclerosis, inflammation may cause other adverse outcomes in kidney disease such as refractory anemia, laboratory signs of iron overload, or a poor quality of life [136,137]. Inflamed dialysis patients frequently display increased serum ferritin, which is a positive acute-phase reactant and an indicator of increased iron burden [138,139]. Serum ferritin levels correlate with hospitalization rates, and a recent increase in serum ferritin concentration in hemodialysis patients may be associated with an enhanced risk of death [138]. (See "Treatment of anemia in nondialysis chronic kidney disease" and "Hyporesponse to erythropoiesis-stimulating agents (ESAs) in chronic kidney disease".)

Some studies, however, have not found an association between malnutrition/inflammation and an increased risk of atherosclerosis among patients on dialysis [140,141]. Among over 50,000 incident Medicare patients on dialysis, for example, there was no association between body mass indices (BMIs) suggestive of poor nutritional status and adverse cardiovascular outcomes [140]. These findings suggest that markers other than BMI, such as a subjective global assessment (SGA), may be preferable when assessing a poor nutritional state in patients on dialysis [141].

MANAGEMENT ISSUES

Overview — Inflammation directly resulting from a specific identified source, such as an ongoing chronic infection, should be aggressively treated with targeted therapy. By comparison, the indications for the use of nonspecific measures to possibly help reduce chronic inflammatory processes are less clear.

Although epidemiologic evidence links inflammation to poor outcome in individuals with kidney function impairment, there are no randomized clinical trials to suggest that outcomes are improved by nonspecific inflammation-reducing modalities. There is therefore no consensus upon a uniform or standardized strategy for the treatment of chronic inflammation in patients with kidney function impairment.

Despite this, the 2005 Kidney Disease Outcomes Quality Initiative (KDOQI) guidelines suggest that the assessment of C-reactive protein (CRP) levels to help assess the presence of underlying inflammation would be beneficial [142]. The presence of increased levels, usually defined as a serum level greater than 5 to 10 mg/L, may help predict cardiovascular risk.

General interventions — The following interventions are beneficial among patients with chronic kidney disease (CKD) or end-stage kidney disease (ESKD) with inflammation:

Evaluate and treat occult infection

Assess for and treat periodontal disease and poor dental hygiene

Screen for tuberculosis

Evaluate and treat urinary tract infections

Routinely assess for pericarditis

Evaluate and treat diabetic foot ulcers

Remove old, nonfunctioning arteriovenous grafts

Remove failed kidney allografts

Replace standard dialysate fluid with ultrapure dialysate

Treat rheumatologic conditions (ie, vasculitis, de novo or recurrent lupus flares)

Targeted pharmacologic therapies — If utilized, treatment modalities may target inflammation directly or ameliorate oxidative stress or endothelial dysfunction. The following agents have been evaluated [143,144]:

Statins – Statins decrease CRP levels independent of their effects on lipids [145-147]. A randomized, placebo-controlled trial of 34 dialysis patients reported that atorvastatin (10 mg/day), after six months, significantly lowered serum CRP (9 and 5 mg/L) and cholesterol concentrations (175 and 156 mg/dL) and increased serum albumin levels (3.69 and 3.99 g/dL) [145]. A meta-analysis of nine clinical trials examining the effect of statin therapy on inflammatory and nutritional markers reported a significant decrease in CRP and high-sensitivity CRP levels but no effect on serum albumin levels [148]. Early data suggested that the use of statins have been associated with decreased mortality [149], but two subsequent, large clinical trials have failed to show a benefit of treatment with atorvastatin [150] and rosuvastatin [151] on clinical endpoints in chronic hemodialysis patients. This was in spite of an 11.5 percent decrease in high-sensitivity CRP levels in patients treated with rosuvastatin in the AURORA study [151], raising doubts about the significance of CRP in cardiovascular morbidity and mortality in this patient population. A third study (SHARP) [152] examined the effects of simvastatin and ezetimibe on clinical endpoints in patients with various stages of CKD (dialysis and nondialysis dependent) and found a significant decrease in a composite cardiovascular event rate in patients treated with simvastatin and ezetimibe [153]. Interestingly, while patients with higher baseline levels of CRP were at increased risk of future cardiovascular and noncardiovascular events in SHARP, the intervention with simvastatin and ezetimibe reduced cardiovascular events independent of CRP level, suggesting that their cardiovascular benefits were not mediated through control of inflammation [154]. (See "Secondary prevention of cardiovascular disease in end-stage kidney disease (dialysis)", section on 'SHARP trial'.)

Renin-angiotensin-aldosterone system blockers – Angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers, and mineralocorticoid receptor antagonists are used as antihypertensive agents and in the treatment of heart failure, but some of their benefits in patients with CKD and heart failure may be related to effects that are independent from their blood pressure-lowering mechanism. One such mechanism is an antiinflammatory effect [155]. Although the data are inconsistent, ACE inhibitors may have antiinflammatory properties in both the general population and in those with CKD [156,157]. Their utility to treat inflammation has not been formally studied in patients with CKD. Finerenone, a nonsteroidal selective mineralocorticoid receptor antagonist, has proven kidney and cardiac benefits [158,159]. The putative mechanisms of action of finerenone are complex and involve effects on inflammation, oxidative stress, and fibrosis [160]. Nevertheless, the impact of finerenone therapy on systemic inflammation in CKD is unclear.

Sodium-glucose cotransporter 2 inhibitors – Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a class of antihyperglycemic agents that lower blood sugar through glycosuric actions [161]. SGLT2 inhibitors have been found to improve kidney and cardiovascular outcomes in diverse patient populations including patients with CKD [162-169]. These benefits appear to be mediated by complex metabolic affects, which include the suppression of inflammation [170-173].

Apabetalone – Apabetalone is a selective modulator of the bromodomain and extraterminal domain transcription system. As an epigenetic modulator, apabetalone has complex mechanisms of action that include (among others) a downregulation of inflammatory responses. Apabetalone has been proposed as a potential therapy for cardiovascular disease in CKD [174]; however, the clinical utility of this agent is uncertain [175,176]. [175,176]

Antioxidants – Since oxidative stress contributes to inflammation in CKD, antioxidants may lower the incidence of cardiovascular events in patients on hemodialysis:

High-dose vitamin E was found to lower the risk of cardiovascular events in a small clinical trial in patients on dialysis [177]. A systematic review and meta-analysis concluded that dialyzers coated with vitamin E can reduce oxidative stress and inflammation [178], although a subsequent randomized, controlled clinical trial found no effect on CRP from vitamin E-coated dialyzer membranes compared with non-vitamin E-coated membranes [179]. The use of vitamin C infusion and of vitamin E-coated dialyzers in hemodialysis patients was also shown to reduce oxidative stress [180].

One small study in dialysis patients showed that treatment with acetylcysteine lowered the incidence of cardiac events [181]. Another small clinical trial in patients on hemodialysis showed a reduction in CRP and interleukin (IL) 6 after a three-month intervention with acetylcysteine [182]. Most studies indicating an antiinflammatory and antioxidant effect of acetylcysteine in patients with CKD or ESKD are experimental, with very few and small clinical trials among them [183].

Dietary supplementation with Brazil nut, a food source rich in selenium, resulted in a significant decrease in oxidative stress and inflammation in small clinical trials of patients with ESKD [184,185]. Other nutraceuticals with antiinflammatory and antioxidant properties in experimental studies and in small clinical trials in patients with CKD or ESKD include dark chocolate, propolis, curcumin, resistant starch, beetroot, and cinnamon. Further examination of these in larger clinical trials will be needed to assess their effects on inflammation and clinical outcomes [186-192].

A meta-analysis of clinical trials that examined the effects of various antioxidant therapies on mortality, cardiovascular, and kidney events failed to detect significant effect on mortality and cardiovascular events overall, but there seemed to be benefits in reducing cardiovascular events in patients on dialysis [193]. The small sample size of most studies and the heterogeneity of the studies included in this meta-analysis make it difficult to generalize its results. It is also unclear to what extent the beneficial effects associated with the use of these antioxidants is related to their antiinflammatory effect. The feasibility and safety of these agents await testing in larger studies.

Sevelamer hydrochlorideSevelamer hydrochloride is a cationic polymer that is used in patients with ESKD as an intestinal phosphate binder. This agent also possesses "pleiotropic" effects that are unrelated to phosphate binding; one such effect may be the amelioration of inflammation. Sevelamer lowered CRP and other markers of inflammation in some [194-196], but not all [197], studies. A systematic review and meta-analysis that examined the pleiotropic effects of sevelamer found that sevelamer lowered glycated hemoglobin and improved lipid profiles but had no significant effect on inflammatory biomarkers [198].

Heparin – Heparin is a glycosaminoglycan used as an anticoagulant and has been recognized for its antiinflammatory properties; this appears to be distinct from its anticoagulant activity [199]. It was proposed that new agents could be developed that retain the antiinflammatory properties of heparin but without the anticoagulant effects [199,200]. In our opinion, the practical impact of a heparin-related antiinflammatory effect in the ESKD population is questionable given that heparin is already routinely applied as an anticoagulant with hemodialysis, yet chronic inflammation is still very common in these patients.

Megestrol acetateMegestrol acetate is a synthetic derivate of progesterone used as an appetite stimulant. This agent also inhibits the activity of IL-1, IL-6, and tumor necrosis factor alpha (TNF-alpha) [201-205]. The beneficial effects of megestrol acetate in patients with ESKD are related to its appetite-stimulant effects and include improved appetite, increased energy and protein intake, and increased dry weight and quality of life [206-208]. The following side effects limit its usefulness: headaches, dizziness, confusion, diarrhea, hyperglycemia, thromboembolic phenomena, breakthrough uterine bleeding, peripheral edema, hypertension, adrenal suppression, and adrenal insufficiency [201]. A small clinical trial of 12 patients on dialysis receiving megestrol acetate described a beneficial effect on appetite and on nutritional markers but no effect on inflammatory markers [209].

Bardoxolone methyl – Bardoxolone methyl (RTA 402) is an antioxidant inflammation modulator (AIM) in clinical development for various inflammation and cancer-related indications. It inhibits immune-mediated inflammation by restoring redox homeostasis in inflamed tissues through the induction of the cytoprotective transcription factor Nrf2 and suppresses the activities of the pro-oxidant and proinflammatory transcription factors nuclear factor kappa B (NF-kB) and the signal transducers and activators of transcription (STATs). In vivo, bardoxolone has shown significant antiinflammatory activity in various animal models of inflammation such as kidney damage in the cisplatin model and ischemia-reperfusion model of acute kidney injury. In a phase II clinical trial, bardoxolone methyl therapy led to a significant increase in estimated glomerular filtration rate (eGFR) levels in patients with advanced diabetic kidney disease [210]. A phase III clinical trial designed to test the hypothesis that bardoxolone methyl decreases the incidence of ESKD was terminated for safety reasons due to volume retention among the participants [211]. In a subsequent trial in which 157 patients with Alport syndrome were randomly assigned to bardoxolone methyl or placebo, eGFR was higher in the bardoxolone methyl group at two prespecified time points [212]. However, treatment with bardoxolone methyl was associated with an increase in urinary albumin excretion and with liver enzyme elevations. In addition, bardoxolone methyl had no effect on progression to kidney failure. Based on these data, the US Food and Drug Administration did not approve bardoxolone methyl for treatment of Alport syndrome.

Omega-3 fatty acids – Omega-3 fatty acids have antiinflammatory effects, and higher dietary intake of omega-3 fatty acids has been associated with less inflammation [213]. However, a 2018 meta-analysis of 12 placebo-controlled trials, including 487 patients with CKD, concluded that omega-3 fatty acid supplementation did not lower levels of the inflammatory markers CRP, IL-6, or TNF-alpha [214]. Another meta-analysis has yielded similar results [215]. However, a 2023 meta-analysis of seven eligible trials found that omega-3 fatty acid therapy was as beneficial as a nonsurgical treatment of periodontal disease [216]. Since periodontal disease may be an important contributor to the inflammation seen in patients with kidney disease, it is possible that omega-3 fatty acids may alleviate inflammation in selected patients with CKD.

Vitamin D – Vitamin D has multiple effects that have been invoked as potential explanations for its association with improved survival in patients with CKD and ESKD [217]. One such effect is modulation of the inflammatory reaction. In a small proof-of-concept clinical trial, the administration of the selective vitamin D receptor activator, paricalcitol, resulted in significant reduction in CRP, IL-6, and TNF-alpha levels in patients with advanced nondialysis-dependent CKD [218]. However, in a double-blind, randomized, placebo-controlled clinical trial including 46 patients with early CKD, high-dose cholecalciferol supplementation did not result in a significant change in most measured markers of inflammation [219]. Furthermore, in a double-blind, randomized, placebo-controlled clinical trial of ergocalciferol supplementation in 276 patients with ESKD on hemodialysis, ergocalciferol supplementation for six months resulted in no significant change in CRP levels (among other outcomes) [220]. A 2022 meta-analysis that included 18 trials with 1834 patients found that vitamin D supplementation did not lower levels of CRP, IL-6, or TNF-alpha in patients with CKD [221].

L-carnitine – L-carnitine deficiency is common in patients on dialysis. A systematic meta-analysis of clinical trials examining the effects of L-carnitine supplementation in hemodialysis patients indicated that this therapy resulted in a significant reduction in CRP levels [222]. A more recent, small randomized trial of L-carnitine supplementation in 24 children on hemodialysis showed a significant reduction in IL-6 levels and a nonsignificant reduction in high-sensitivity CRP levels in patients treated with L-carnitine [223].

Probiotic therapy – The intestinal microbiome is significantly altered among patients with CKD and is emerging as an important risk factor in the development and progression of kidney and heart disease [224,225]. Probiotics have emerged as a potential therapy with broad benefits in patients with CKD and ESKD [226,227].A 2018 meta-analysis of randomized trials (423 patients) including patients with CKD concluded that treatment with probiotic therapy reduced formation of the uremic solute p-cresol, which has been implicated in oxidative stress and systemic inflammation in the setting of CKD [228]. A subsequent meta-analysis of randomized trials involving patients on dialysis (178 patients) found that treatment with probiotics lowered CRP levels, but not TNF-alpha and albumin levels [229]. Broader dietary interventions such as plant-based diets and others remain promising alternatives to positively affect the intestinal microbiome and to alleviate the negative consequences of the uremic milieu [230-234]. The efficacy and safety of these will need to be tested in clinical trials.

In addition to the above agents and interventions with complex mechanisms of action, there are multiple "designer" drugs that target a specific aspect of the inflammatory cascade. These drugs have applications mainly in rheumatoid arthritis, and most of them will likely not be utilized in patients with CKD. However, some agents are considered for patients with CKD:

EtanerceptEtanercept is an anti-TNF agent that is effective in the treatment of various forms of inflammatory arthritis, such as rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis. In a pilot study involving 10 patients on chronic hemodialysis, the administration of etanercept over 44 weeks appeared safe but resulted only in small improvement in prealbumin concentration [235]. A phase II, randomized, double-blind, placebo-controlled clinical trial examining the effectiveness of etanercept in improving serum albumin and CRP levels of patients on hemodialysis was terminated due to insufficient patient enrollment [236].

PentoxifyllinePentoxifylline is a nonspecific phosphodiesterase inhibitor that inhibits TNF transcription [237]. Pentoxifylline has been tested in patients with ESKD in a randomized, double-blinded, placebo-controlled clinical trial [238]. The Anti-Inflammatory and Anti-Oxidative Nutrition in Dialysis Patients (AIONID) study was sponsored by the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) and examined the effect of oral nutritional supplements with antiinflammatory and antioxidant properties along with pentoxifylline therapy in a factorial design on malnutrition and inflammation in 100 patients receiving maintenance hemodialysis. Daily intake of a high-protein nutritional supplement with antiinflammatory and antioxidative ingredients resulted in a slight but significant increase in serum albumin levels.

Other studies that tested the efficacy of pentoxifylline were less successful in alleviating inflammation. In a small clinical trial of 71 adult patients with elevated CRP, the administration of 400 mg of pentoxifylline thrice weekly did not result in significant changes in serum hepcidin level compared with placebo [239]. This study did not test the effects of pentoxifylline on markers of inflammation such as CRP. Early-phase clinical trials are examining the pentoxifylline derivative CTP-499 as a treatment in diabetic kidney disease [240,241].

IL-1 antagonistsAnakinra is a human recombinant IL-1 receptor antagonist that is used in rheumatoid arthritis. It also shows some promise in the treatment of inflammation in patients with CKD. A single study examined the pharmacokinetics of this drug in patients with impaired kidney function, showing that the main route of elimination for anakinra is renal clearance and that hemodialysis has a very small effect on clearance [242]. The results of this study indicate that thrice-weekly dosing of anakinra may be possible. The safety and efficacy of anakinra in chronic hemodialysis patients was examined in a randomized, controlled trial. This study reported significant beneficial effects of anakinra on serum CRP and IL-6 levels but no effects on serum albumin, prealbumin, and lean body mass levels [243]. Similarly, another trial that randomly assigned 42 patients with stage 3 and 4 CKD to receive rilonacept (another IL-1 receptor antagonist) or placebo found lower high-sensitivity CRP levels in patients receiving rilonacept [244]. Canakinumab is an IL-1-beta antagonist that lowered IL-6 and high-sensitivity CRP levels in CANTOS (Canakinumab Anti-inflammatory Thrombosis Outcomes Study), which enrolled 10,061 patients with diabetes and systemic inflammation (high-sensitivity CRP >2 mg/mL) [245]. In a post-hoc analysis of CANTOS, patients with early-stage CKD (eGFR <60 mL/min/1.73m2) treated with canakinumab (versus placebo) showed a significant reduction in major adverse cardiovascular events. This effect was largest in patients who showed a robust antiinflammatory effect after the first dose of canakinumab [246].

IL-6 antagonists – Ziltivekimab is a novel anti-IL-6 ligand antibody that reduced CRP levels in a clinical trial of 264 patients with moderate to severe CKD and high-sensitivity CRP >2 mg/L [247]. Similar dose-dependent reductions were seen for fibrinogen, serum amyloid A, haptoglobin, secretory phospholipase A2, and lipoprotein(a). Ziltivekimab also reduced inflammatory biomarker levels and erythropoiesis-stimulating agent (ESA) requirements in a phase I/II clinical trial conducted in patients on hemodialysis with inflammation and ESA hyporesponsiveness [248]. Another small trial in Japan randomly assigned 36 patients on chronic hemodialysis to two different doses of ziltivekimab or placebo [249]. Compared with patients treated with placebo, patients treated with ziltivekimab showed significant reductions in inflammatory biomarkers after 12 weeks of therapy. Larger clinical trials are needed to test the efficacy and safety of this agent and its impact on clinical outcomes.

RituximabRituximab is a B-cell-depleting monoclonal anti-CD20 antibody [250]. B-cell depletion affects the levels of autoantibodies that play important roles in the pathophysiology of diseases such as rheumatoid factor in rheumatoid arthritis [251,252], systemic lupus erythematosus [253], and antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis [254-256]. Rituximab also has been examined for the treatment of various glomerular diseases [257-261]. It is unclear if this agent will be employed for the treatment of chronic inflammation in CKD since the autoantibody depletion resultant after B-cell depletion may be more specific to certain types of diseases.

Chemokine inhibitors – Several agents targeting chemokine pathways have been evaluated for the treatment of diabetic nephropathy, such as C-C chemokine receptor type 2 (CCR2) inhibitors [262], the C-C motif-ligand 2 (CCL2) (ie, monocyte chemotactic protein 1) inhibitor emapticap pegol [263], or the Janus kinase 1 (JAK1)/Janus kinase 2 (JAK2) inhibitor baricitinib [264]. (See "Treatment of diabetic kidney disease".)

Hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF PHIs) – This class of agents is used for the treatment of anemia in ESKD. HIF PHIs may be more effective in treating anemia associated with inflammation, which is typically resistant to traditional anemia therapies [265,266]. Preclinical studies suggest that HIF PHIs may also directly decrease inflammation [267].

Optimization of dialysis treatment may also improve inflammatory status in patients on dialysis, with the type of dialysis membrane and dialysis method (online hemodiafiltration versus standard hemodialysis) possibly having an impact upon mortality [268,269]. As an example, the use of biocompatible membranes decreases serum levels of CRP, although such interventions may not completely normalize its serum concentration [66,67]. (See "Clinical consequences of hemodialysis membrane biocompatibility".)

However, a major clinical trial, known as the HEMO study, failed to show a major improvement in clinical outcomes with the use of high-flux membranes. Nevertheless, this finding does not necessarily refute a role of dialysis membranes in the development of chronic inflammation in patients with ESKD, since the membrane effect on survival may have been small compared with that of other variables.

There is no consensus as to which medication or combination of drugs should be used under different clinical conditions. In general, ACE inhibitors are commonly used in patients with CKD or ESKD with hypertension and/or heart failure, statins are used in those with hyperlipidemia, and vitamin E in patients with refractory muscle cramps. Clinical trials have not been performed to determine whether the use of these medications is warranted for the treatment of inflammation in patients with kidney dysfunction but without an indicated concurrent comorbid condition.

NOVEL APPROACHES — Novel treatment strategies are needed to help manage inflammation among patients with kidney disease:

Additional clinical trials of antiinflammatory and anticytokine agents should be conducted in patients with CKD and ESKD [174].  

Clinical trials are needed to compare the effect of antiinflammatory agents with that of nutritional support in patients suffering from the malnutrition-inflammation complex syndrome (MICS) [112,143].

Limited evidence suggests that resistance training may help reduce MICS and improve cardiometabolic profiles in patients with CKD and ESKD [270-274].

SUMMARY AND RECOMMENDATIONS

Inflammation and kidney disease – Chronic inflammatory processes are common in individuals with chronic kidney disease (CKD), including those with early-stage CKD and especially end-stage kidney disease (ESKD). This is due to many underlying factors, including the uremic milieu, elevated levels of circulating proinflammatory cytokines, oxidative stress, carbonyl stress, protein-energy wasting (PEW), enhanced incidence of infections (especially dialysis access related), and others. (See 'Introduction' above and 'Inflammation and kidney disease' above.)

General markers of inflammation – Routine monitoring of inflammatory markers in all patients with ESKD or CKD is not recommended by most guidelines, although it may improve patient management, for example, by providing evidence of an unrecognized infection or other treatable inflammatory conditions. Periodic measurement of C-reactive protein (CRP) or interleukin (IL) 6 to assess severity and frequency of inflammation is reasonable in select patients as part of an individualized work-up prompted by specific clinical characteristics, such as PEW. (See 'General markers of inflammation' above.)

Management issues – The presence of elevated inflammatory markers (eg, CRP >5 mg/L or 0.5 mg/dL) in patients with ESKD or CKD should prompt a search for immediately treatable causes of acute or chronic inflammation, such as occult infections, volume overload, inadequate dialysis treatment, PEW, or the presence of a nonfunctioning kidney allograft. (See 'Management issues' above.)

Novel approaches – No pharmacologic interventions are currently recommended to specifically target chronic inflammation in patients with CKD or ESKD. However, pharmacologic and nutritional interventions may become available in the future for this indication. (See 'Management issues' above and 'Novel approaches' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Joel Kopple, MD, who contributed to an earlier version of this topic review.

  1. 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.
  2. Owen WF, Lowrie EG. C-reactive protein as an outcome predictor for maintenance hemodialysis patients. Kidney Int 1998; 54:627.
  3. Zimmermann J, Herrlinger S, Pruy A, et al. Inflammation enhances cardiovascular risk and mortality in hemodialysis patients. Kidney Int 1999; 55:648.
  4. Zoccali C, Benedetto FA, Mallamaci F, et al. Inflammation is associated with carotid atherosclerosis in dialysis patients. Creed Investigators. Cardiovascular Risk Extended Evaluation in Dialysis Patients. J Hypertens 2000; 18:1207.
  5. Iseki K, Tozawa M, Yoshi S, Fukiyama K. Serum C-reactive protein (CRP) and risk of death in chronic dialysis patients. Nephrol Dial Transplant 1999; 14:1956.
  6. Noh H, Lee SW, Kang SW, et al. Serum C-reactive protein: a predictor of mortality in continuous ambulatory peritoneal dialysis patients. Perit Dial Int 1998; 18:387.
  7. Kalantar-Zadeh K. Recent advances in understanding the malnutrition-inflammation-cachexia syndrome in chronic kidney disease patients: What is next? Semin Dial 2005; 18:365.
  8. Kalantar-Zadeh K. Inflammatory marker mania in chronic kidney disease: pentraxins at the crossroad of universal soldiers of inflammation. Clin J Am Soc Nephrol 2007; 2:872.
  9. Qureshi AR, Alvestrand A, Divino-Filho JC, et al. Inflammation, malnutrition, and cardiac disease as predictors of mortality in hemodialysis patients. J Am Soc Nephrol 2002; 13 Suppl 1:S28.
  10. Kaysen GA, Dubin JA, Müller HG, et al. Relationships among inflammation nutrition and physiologic mechanisms establishing albumin levels in hemodialysis patients. Kidney Int 2002; 61:2240.
  11. Kalantar-Zadeh K, Kopple JD. Relative contributions of nutrition and inflammation to clinical outcome in dialysis patients. Am J Kidney Dis 2001; 38:1343.
  12. Kurts C, Panzer U, Anders HJ, Rees AJ. The immune system and kidney disease: basic concepts and clinical implications. Nat Rev Immunol 2013; 13:738.
  13. Bemelmans MH, Gouma DJ, Buurman WA. Influence of nephrectomy on tumor necrosis factor clearance in a murine model. J Immunol 1993; 150:2007.
  14. Poole S, Bird TA, Selkirk S, et al. Fate of injected interleukin 1 in rats: sequestration and degradation in the kidney. Cytokine 1990; 2:416.
  15. Panichi V, Migliori M, De Pietro S, et al. C reactive protein in patients with chronic renal diseases. Ren Fail 2001; 23:551.
  16. Stenvinkel P, Heimbürger O, Wang T, et al. High serum hyaluronan indicates poor survival in renal replacement therapy. Am J Kidney Dis 1999; 34:1083.
  17. Guessous I, Ponte B, Marques-Vidal P, et al. Clinical and biological determinants of kidney outcomes in a population-based cohort study. Kidney Blood Press Res 2014; 39:74.
  18. Puthumana J, Thiessen-Philbrook H, Xu L, et al. Biomarkers of inflammation and repair in kidney disease progression. J Clin Invest 2021; 131.
  19. Chung SH, Heimbürger O, Stenvinkel P, et al. Association between inflammation and changes in residual renal function and peritoneal transport rate during the first year of dialysis. Nephrol Dial Transplant 2001; 16:2240.
  20. Pecoits-Filho R, Heimbürger O, Bárány P, et al. Associations between circulating inflammatory markers and residual renal function in CRF patients. Am J Kidney Dis 2003; 41:1212.
  21. Eleftheriadis T, Pissas G, Antoniadi G, et al. Damage-associated molecular patterns derived from mitochondria may contribute to the hemodialysis-associated inflammation. Int Urol Nephrol 2014; 46:107.
  22. Sumida K, Kovesdy CP. The gut-kidney-heart axis in chronic kidney disease. Physiol Int 2019; 106:195.
  23. Hasper D, Hummel M, Kleber FX, et al. Systemic inflammation in patients with heart failure. Eur Heart J 1998; 19:761.
  24. Sato Y, Takatsu Y, Kataoka K, et al. Serial circulating concentrations of C-reactive protein, interleukin (IL)-4, and IL-6 in patients with acute left heart decompensation. Clin Cardiol 1999; 22:811.
  25. Sharma R, Bolger AP, Li W, et al. Elevated circulating levels of inflammatory cytokines and bacterial endotoxin in adults with congenital heart disease. Am J Cardiol 2003; 92:188.
  26. Shi K, Wang F, Jiang H, et al. Gut bacterial translocation may aggravate microinflammation in hemodialysis patients. Dig Dis Sci 2014; 59:2109.
  27. Locatelli F, Canaud B, Eckardt KU, et al. The importance of diabetic nephropathy in current nephrological practice. Nephrol Dial Transplant 2003; 18:1716.
  28. Himmelfarb J, Stenvinkel P, Ikizler TA, Hakim RM. The elephant in uremia: oxidant stress as a unifying concept of cardiovascular disease in uremia. Kidney Int 2002; 62:1524.
  29. Spittle MA, Hoenich NA, Handelman GJ, et al. Oxidative stress and inflammation in hemodialysis patients. Am J Kidney Dis 2001; 38:1408.
  30. Mezzano D, Pais EO, Aranda E, et al. Inflammation, not hyperhomocysteinemia, is related to oxidative stress and hemostatic and endothelial dysfunction in uremia. Kidney Int 2001; 60:1844.
  31. Stenvinkel P, Holmberg I, Heimbürger O, Diczfalusy U. A study of plasmalogen as an index of oxidative stress in patients with chronic renal failure. Evidence of increased oxidative stress in malnourished patients. Nephrol Dial Transplant 1998; 13:2594.
  32. Miyata T, Ueda Y, Horie K, et al. Renal catabolism of advanced glycation end products: the fate of pentosidine. Kidney Int 1998; 53:416.
  33. Suliman ME, Heimbürger O, Bárány P, et al. Plasma pentosidine is associated with inflammation and malnutrition in end-stage renal disease patients starting on dialysis therapy. J Am Soc Nephrol 2003; 14:1614.
  34. Shimizu H, Bolati D, Adijiang A, et al. NF-κB plays an important role in indoxyl sulfate-induced cellular senescence, fibrotic gene expression, and inhibition of proliferation in proximal tubular cells. Am J Physiol Cell Physiol 2011; 301:C1201.
  35. Kalantar-Zadeh K, Kopple JD, Deepak S, et al. Food intake characteristics of hemodialysis patients as obtained by food frequency questionnaire. J Ren Nutr 2002; 12:17.
  36. Langlois M, Duprez D, Delanghe J, et al. Serum vitamin C concentration is low in peripheral arterial disease and is associated with inflammation and severity of atherosclerosis. Circulation 2001; 103:1863.
  37. Deicher R, Ziai F, Bieglmayer C, et al. Low total vitamin C plasma level is a risk factor for cardiovascular morbidity and mortality in hemodialysis patients. J Am Soc Nephrol 2005; 16:1811.
  38. Yamazaki H, Tanji K, Wakabayashi K, et al. Role of the Keap1/Nrf2 pathway in neurodegenerative diseases. Pathol Int 2015; 65:210.
  39. Choi BH, Kang KS, Kwak MK. Effect of redox modulating NRF2 activators on chronic kidney disease. Molecules 2014; 19:12727.
  40. Cardozo LF, Pedruzzi LM, Stenvinkel P, et al. Nutritional strategies to modulate inflammation and oxidative stress pathways via activation of the master antioxidant switch Nrf2. Biochimie 2013; 95:1525.
  41. Howden R. Nrf2 and cardiovascular defense. Oxid Med Cell Longev 2013; 2013:104308.
  42. 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.
  43. Stenvinkel P, Painer J, Kuro-O M, et al. Novel treatment strategies for chronic kidney disease: insights from the animal kingdom. Nat Rev Nephrol 2018; 14:265.
  44. Pedruzzi LM, Cardozo LF, Daleprane JB, et al. Systemic inflammation and oxidative stress in hemodialysis patients are associated with down-regulation of Nrf2. J Nephrol 2015; 28:495.
  45. Keane WF, Collins AJ. Influence of co-morbidity on mortality and morbidity in patients treated with hemodialysis. Am J Kidney Dis 1994; 24:1010.
  46. Kshirsagar AV, Moss KL, Elter JR, et al. Periodontal disease is associated with renal insufficiency in the Atherosclerosis Risk In Communities (ARIC) study. Am J Kidney Dis 2005; 45:650.
  47. Kaysen GA. C-reactive protein: a story half told. Semin Dial 2000; 13:143.
  48. Sarnak MJ, Jaber BL. Mortality caused by sepsis in patients with end-stage renal disease compared with the general population. Kidney Int 2000; 58:1758.
  49. Ishigami J, Matsushita K. Clinical epidemiology of infectious disease among patients with chronic kidney disease. Clin Exp Nephrol 2019; 23:437.
  50. Ishigami J, Grams ME, Chang AR, et al. CKD and Risk for Hospitalization With Infection: The Atherosclerosis Risk in Communities (ARIC) Study. Am J Kidney Dis 2017; 69:752.
  51. Lau WL, Kalantar-Zadeh K, Vaziri ND. The Gut as a Source of Inflammation in Chronic Kidney Disease. Nephron 2015; 130:92.
  52. Sumida K, Molnar MZ, Potukuchi PK, et al. Constipation and Incident CKD. J Am Soc Nephrol 2017; 28:1248.
  53. Sumida K, Yamagata K, Kovesdy CP. Constipation in CKD. Kidney Int Rep 2020; 5:121.
  54. Sumida K, Molnar MZ, Potukuchi PK, et al. Constipation and risk of death and cardiovascular events. Atherosclerosis 2019; 281:114.
  55. Lin TY, Hung SC. Association of subjective global assessment of nutritional status with gut microbiota in hemodialysis patients: a case-control study. Nephrol Dial Transplant 2021; 36:1104.
  56. Pecoits-Filho R, Stenvinkel P, Marchlewska A, et al. A functional variant of the myeloperoxidase gene is associated with cardiovascular disease in end-stage renal disease patients. Kidney Int Suppl 2003; :S172.
  57. Chen LP, Chiang CK, Chan CP, et al. Does periodontitis reflect inflammation and malnutrition status in hemodialysis patients? Am J Kidney Dis 2006; 47:815.
  58. Craig RG, Kotanko P, Kamer AR, Levin NW. Periodontal diseases--a modifiable source of systemic inflammation for the end-stage renal disease patient on haemodialysis therapy? Nephrol Dial Transplant 2007; 22:312.
  59. Niedzielska I, Chudek J, Kowol I, et al. The odontogenic-related microinflammation in patients with chronic kidney disease. Ren Fail 2014; 36:883.
  60. Sumida K, Han Z, Dashputre AA, et al. Association between Nrf2 and CDKN2A expression in patients with end-stage renal disease: a pilot study. Aging (Albany NY) 2020; 12:16357.
  61. Docherty MH, O'Sullivan ED, Bonventre JV, Ferenbach DA. Cellular Senescence in the Kidney. J Am Soc Nephrol 2019; 30:726.
  62. Sumida K, Mozhui K, Liang X, et al. Association of DNA methylation signatures with premature ageing and cardiovascular death in patients with end-stage kidney disease: a pilot epigenome-wide association study. Epigenetics 2023; 18:2214394.
  63. Carmona A, Guerrero F, Jimenez MJ, et al. Inflammation, Senescence and MicroRNAs in Chronic Kidney Disease. Front Cell Dev Biol 2020; 8:739.
  64. Stenvinkel P. New insights on inflammation in chronic kidney disease-genetic and non-genetic factors. Nephrol Ther 2006; 2:111.
  65. Huang Z, Vlasschaert C, Robinson-Cohen C, et al. Emerging evidence on the role of clonal hematopoiesis of indeterminate potential in chronic kidney disease. Transl Res 2023; 256:87.
  66. Schindler R, Boenisch O, Fischer C, Frei U. Effect of the hemodialysis membrane on the inflammatory reaction in vivo. Clin Nephrol 2000; 53:452.
  67. Memoli B, Minutolo R, Bisesti V, et al. Changes of serum albumin and C-reactive protein are related to changes of interleukin-6 release by peripheral blood mononuclear cells in hemodialysis patients treated with different membranes. Am J Kidney Dis 2002; 39:266.
  68. Ayus JC, Sheikh-Hamad D. Silent infection in clotted hemodialysis access grafts. J Am Soc Nephrol 1998; 9:1314.
  69. Cappelli G, Tetta C, Canaud B. Is biofilm a cause of silent chronic inflammation in haemodialysis patients? A fascinating working hypothesis. Nephrol Dial Transplant 2005; 20:266.
  70. Dukkipati R, Molnar MZ, Park J, et al. Association of vascular access type with inflammatory marker levels in maintenance hemodialysis patients. Semin Dial 2014; 27:415.
  71. Hung A, Pupim L, Yu C, et al. Determinants of C-reactive protein in chronic hemodialysis patients: relevance of dialysis catheter utilization. Hemodial Int 2008; 12:236.
  72. Banerjee T, Kim SJ, Astor B, et al. Vascular access type, inflammatory markers, and mortality in incident hemodialysis patients: the Choices for Healthy Outcomes in Caring for End-Stage Renal Disease (CHOICE) Study. Am J Kidney Dis 2014; 64:954.
  73. Sabry AA, Elshafey EM, Alsaran K, et al. The level of C-reactive protein in chronic hemodialysis patients: a comparative study between patients with noninfected catheters and arteriovenous fistula in two large Gulf hemodialysis centers. Hemodial Int 2014; 18:674.
  74. Pecoits-Filho R, Stenvinkel P, Wang AY, et al. Chronic inflammation in peritoneal dialysis: the search for the holy grail? Perit Dial Int 2004; 24:327.
  75. Avram MM, Fein PA, Rafiz MA, et al. Malnutrition and inflammation as predictors of mortality in peritoneal dialysis patients. Kidney Int 2006; 70(Suppl 104):S4.
  76. Yeun JY, Kaysen GA. Acute phase proteins and peritoneal dialysate albumin loss are the main determinants of serum albumin in peritoneal dialysis patients. Am J Kidney Dis 1997; 30:923.
  77. Palomo-Piñón S, Mora-Villalpando CJ, Del Carmen Prado-Uribe M, et al. Inflammation and myocardial damage markers influence loss of residual renal function in peritoneal dialysis patients. Arch Med Res 2014; 45:484.
  78. López-Gómez JM, Pérez-Flores I, Jofré R, et al. Presence of a failed kidney transplant in patients who are on hemodialysis is associated with chronic inflammatory state and erythropoietin resistance. J Am Soc Nephrol 2004; 15:2494.
  79. Akagun T, Caliskan Y, Yazici H, et al. Elevated resistin levels are associated with inflammation in hemodialysis patients with failed renal allografts. Int J Artif Organs 2014; 37:358.
  80. Kimmel PL, Phillips TM, Simmens SJ, et al. Immunologic function and survival in hemodialysis patients. Kidney Int 1998; 54:236.
  81. Streetz KL, Wüstefeld T, Klein C, et al. Mediators of inflammation and acute phase response in the liver. Cell Mol Biol (Noisy-le-grand) 2001; 47:661.
  82. Suffredini AF, Fantuzzi G, Badolato R, et al. New insights into the biology of the acute phase response. J Clin Immunol 1999; 19:203.
  83. Kalantar-Zadeh K, Kleiner M, Dunne E, et al. Total iron-binding capacity-estimated transferrin correlates with the nutritional subjective global assessment in hemodialysis patients. Am J Kidney Dis 1998; 31:263.
  84. Kaysen GA, Dubin JA, Müller HG, et al. The acute-phase response varies with time and predicts serum albumin levels in hemodialysis patients. The HEMO Study Group. Kidney Int 2000; 58:346.
  85. Stenvinkel P, Barany P, Heimbürger O, et al. Mortality, malnutrition, and atherosclerosis in ESRD: what is the role of interleukin-6? Kidney Int Suppl 2002; :103.
  86. Ketteler M, Bongartz P, Westenfeld R, et al. Association of low fetuin-A (AHSG) concentrations in serum with cardiovascular mortality in patients on dialysis: a cross-sectional study. Lancet 2003; 361:827.
  87. Panichi V, Maggiore U, Taccola D, et al. Interleukin-6 is a stronger predictor of total and cardiovascular mortality than C-reactive protein in haemodialysis patients. Nephrol Dial Transplant 2004; 19:1154.
  88. Kalantar-Zadeh K, Brennan ML, Hazen SL. Serum myeloperoxidase and mortality in maintenance hemodialysis patients. Am J Kidney Dis 2006; 48:59.
  89. Tong M, Carrero JJ, Qureshi AR, et al. Plasma pentraxin 3 in patients with chronic kidney disease: associations with renal function, protein-energy wasting, cardiovascular disease, and mortality. Clin J Am Soc Nephrol 2007; 2:889.
  90. Muntinghe FL, Verduijn M, Zuurman MW, et al. CCR5 deletion protects against inflammation-associated mortality in dialysis patients. J Am Soc Nephrol 2009; 20:1641.
  91. Ducloux D, Bresson-Vautrin C, Kribs M, et al. C-reactive protein and cardiovascular disease in peritoneal dialysis patients. Kidney Int 2002; 62:1417.
  92. Pecoits-Filho R, Bárány P, Lindholm B, et al. Interleukin-6 is an independent predictor of mortality in patients starting dialysis treatment. Nephrol Dial Transplant 2002; 17:1684.
  93. Wang AY, Woo J, Lam CW, et al. Is a single time point C-reactive protein predictive of outcome in peritoneal dialysis patients? J Am Soc Nephrol 2003; 14:1871.
  94. Kalantar-Zadeh K, Kopple JD, Humphreys MH, Block G. Comparing outcome predictability of markers of malnutrition-inflammation complex syndrome in haemodialysis patients. Nephrol Dial Transplant 2004; 19:1507.
  95. Wang AY, Wang M, Woo J, et al. Inflammation, residual kidney function, and cardiac hypertrophy are interrelated and combine adversely to enhance mortality and cardiovascular death risk of peritoneal dialysis patients. J Am Soc Nephrol 2004; 15:2186.
  96. Rao M, Guo D, Perianayagam MC, et al. Plasma interleukin-6 predicts cardiovascular mortality in hemodialysis patients. Am J Kidney Dis 2005; 45:324.
  97. Pecoits-Filho R, Lindholm B, Axelsson J, Stenvinkel P. Update on interleukin-6 and its role in chronic renal failure. Nephrol Dial Transplant 2003; 18:1042.
  98. Rambod M, Kovesdy CP, Kalantar-Zadeh K. Combined high serum ferritin and low iron saturation in hemodialysis patients: the role of inflammation. Clin J Am Soc Nephrol 2008; 3:1691.
  99. Kalantar-Zadeh K, Regidor DL, McAllister CJ, et al. Time-dependent associations between iron and mortality in hemodialysis patients. J Am Soc Nephrol 2005; 16:3070.
  100. Fouque D, Kalantar-Zadeh K, Kopple J, et al. A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease. Kidney Int 2008; 73:391.
  101. Kopple JD. Nutritional status as a predictor of morbidity and mortality in maintenance dialysis patients. ASAIO J 1997; 43:246.
  102. Kalantar-Zadeh K, Block G, Humphreys MH, Kopple JD. Reverse epidemiology of cardiovascular risk factors in maintenance dialysis patients. Kidney Int 2003; 63:793.
  103. Kalantar-Zadeh K, Abbott KC, Kronenberg F, et al. Epidemiology of dialysis patients and heart failure patients. Semin Nephrol 2006; 26:118.
  104. Liu Y, Coresh J, Eustace JA, et al. Association between cholesterol level and mortality in dialysis patients: role of inflammation and malnutrition. JAMA 2004; 291:451.
  105. Flores EA, Bistrian BR, Pomposelli JJ, et al. Infusion of tumor necrosis factor/cachectin promotes muscle catabolism in the rat. A synergistic effect with interleukin 1. J Clin Invest 1989; 83:1614.
  106. Kaizu Y, Kimura M, Yoneyama T, et al. Interleukin-6 may mediate malnutrition in chronic hemodialysis patients. Am J Kidney Dis 1998; 31:93.
  107. Kopple JD, Greene T, Chumlea WC, et al. Relationship between nutritional status and the glomerular filtration rate: results from the MDRD study. Kidney Int 2000; 57:1688.
  108. Kaysen GA, Chertow GM, Adhikarla R, et al. Inflammation and dietary protein intake exert competing effects on serum albumin and creatinine in hemodialysis patients. Kidney Int 2001; 60:333.
  109. Kaysen GA, Dubin JA, Müller HG, et al. Inflammation and reduced albumin synthesis associated with stable decline in serum albumin in hemodialysis patients. Kidney Int 2004; 65:1408.
  110. Bologa RM, Levine DM, Parker TS, et al. Interleukin-6 predicts hypoalbuminemia, hypocholesterolemia, and mortality in hemodialysis patients. Am J Kidney Dis 1998; 32:107.
  111. Kalantar-Zadeh K, Kopple JD, Block G, Humphreys MH. A malnutrition-inflammation score is correlated with morbidity and mortality in maintenance hemodialysis patients. Am J Kidney Dis 2001; 38:1251.
  112. Kalantar-Zadeh K, Ikizler TA, Block G, et al. Malnutrition-inflammation complex syndrome in dialysis patients: causes and consequences. Am J Kidney Dis 2003; 42:864.
  113. Stenvinkel P, Heimbürger O, Lindholm B, et al. Are there two types of malnutrition in chronic renal failure? Evidence for relationships between malnutrition, inflammation and atherosclerosis (MIA syndrome). Nephrol Dial Transplant 2000; 15:953.
  114. Molnar MZ, Carrero JJ, Mucsi I, et al. Comparison of the malnutrition-inflammation score in chronic kidney disease patients and kidney transplant recipients. Int Urol Nephrol 2015; 47:1025.
  115. Ikizler TA, Burrowes JD, Byham-Gray LD, et al. KDOQI Clinical Practice Guideline for Nutrition in CKD: 2020 Update. Am J Kidney Dis 2020; 76:S1.
  116. Pupim LB, Caglar K, Hakim RM, et al. Uremic malnutrition is a predictor of death independent of inflammatory status. Kidney Int 2004; 66:2054.
  117. Kaysen GA, Stevenson FT, Depner TA. Determinants of albumin concentration in hemodialysis patients. Am J Kidney Dis 1997; 29:658.
  118. Ginn HE, Frost A, Lacy WW. Nitrogen balance in hemodialysis patients. Am J Clin Nutr 1968; 21:385.
  119. Leon JB, Majerle AD, Soinski JA, et al. Can a nutrition intervention improve albumin levels among hemodialysis patients? A pilot study. J Ren Nutr 2001; 11:9.
  120. Leon JB, Albert JM, Gilchrist G, et al. Improving albumin levels among hemodialysis patients: a community-based randomized controlled trial. Am J Kidney Dis 2006; 48:28.
  121. Hulsewé KW, van Acker BA, von Meyenfeldt MF, Soeters PB. Nutritional depletion and dietary manipulation: effects on the immune response. World J Surg 1999; 23:536.
  122. De Simone C, Famularo G, Tzantzoglou S, et al. Carnitine depletion in peripheral blood mononuclear cells from patients with AIDS: effect of oral L-carnitine. AIDS 1994; 8:655.
  123. Rauchhaus M, Coats AJ, Anker SD. The endotoxin-lipoprotein hypothesis. Lancet 2000; 356:930.
  124. deFilippi C, Wasserman S, Rosanio S, et al. Cardiac troponin T and C-reactive protein for predicting prognosis, coronary atherosclerosis, and cardiomyopathy in patients undergoing long-term hemodialysis. JAMA 2003; 290:353.
  125. Bazeley J, Bieber B, Li Y, et al. C-reactive protein and prediction of 1-year mortality in prevalent hemodialysis patients. Clin J Am Soc Nephrol 2011; 6:2452.
  126. Stenvinkel P, Heimbürger O, Jogestrand T. Elevated interleukin-6 predicts progressive carotid artery atherosclerosis in dialysis patients: association with Chlamydia pneumoniae seropositivity. Am J Kidney Dis 2002; 39:274.
  127. Sarnak MJ, Coronado BE, Greene T, et al. Cardiovascular disease risk factors in chronic renal insufficiency. Clin Nephrol 2002; 57:327.
  128. Chauveau P, Chadefaux B, Coudé M, et al. Hyperhomocysteinemia, a risk factor for atherosclerosis in chronic uremic patients. Kidney Int Suppl 1993; 41:S72.
  129. Oberg BP, McMenamin E, Lucas FL, et al. Increased prevalence of oxidant stress and inflammation in patients with moderate to severe chronic kidney disease. Kidney Int 2004; 65:1009.
  130. Descamps-Latscha B, Witko-Sarsat V, Nguyen-Khoa T, et al. Advanced oxidation protein products as risk factors for atherosclerotic cardiovascular events in nondiabetic predialysis patients. Am J Kidney Dis 2005; 45:39.
  131. Kovesdy CP, George SM, Anderson JE, Kalantar-Zadeh K. Outcome predictability of biomarkers of protein-energy wasting and inflammation in moderate and advanced chronic kidney disease. Am J Clin Nutr 2009; 90:407.
  132. Gupta J, Dominic EA, Fink JC, et al. Association between Inflammation and Cardiac Geometry in Chronic Kidney Disease: Findings from the CRIC Study. PLoS One 2015; 10:e0124772.
  133. Amdur RL, Feldman HI, Gupta J, et al. Inflammation and Progression of CKD: The CRIC Study. Clin J Am Soc Nephrol 2016; 11:1546.
  134. Kovesdy CP, Kalantar-Zadeh K. Do genes allow inflammation to kill or not to kill? J Am Soc Nephrol 2009; 20:1429.
  135. Muntinghe FL, Verduijn M, Zuurman MW, et al. CCR5 deletion protects against inflammation-associated mortality in dialysis patients. J Am Soc Nephrol 2009; 20:1641.
  136. Kalantar-Zadeh K, Kopple JD, Block G, Humphreys MH. Association among SF36 quality of life measures and nutrition, hospitalization, and mortality in hemodialysis. J Am Soc Nephrol 2001; 12:2797.
  137. Kalantar-Zadeh K, McAllister CJ, Lehn RS, et al. Effect of malnutrition-inflammation complex syndrome on EPO hyporesponsiveness in maintenance hemodialysis patients. Am J Kidney Dis 2003; 42:761.
  138. Kalantar-Zadeh K, Don BR, Rodriguez RA, Humphreys MH. Serum ferritin is a marker of morbidity and mortality in hemodialysis patients. Am J Kidney Dis 2001; 37:564.
  139. Gunnell J, Yeun JY, Depner TA, Kaysen GA. Acute-phase response predicts erythropoietin resistance in hemodialysis and peritoneal dialysis patients. Am J Kidney Dis 1999; 33:63.
  140. Beddhu S, Pappas LM, Ramkumar N, Samore MH. Malnutrition and atherosclerosis in dialysis patients. J Am Soc Nephrol 2004; 15:733.
  141. Stenvinkel P, Heimbürger O, Lindholm B. Wasting, but not malnutrition, predicts cardiovascular mortality in end-stage renal disease. Nephrol Dial Transplant 2004; 19:2181.
  142. K/DOQI Workgroup. K/DOQI clinical practice guidelines for cardiovascular disease in dialysis patients. Am J Kidney Dis 2005; 45:S1.
  143. Guarnieri G, Biolo G, Zanetti M, Barazzoni R. Chronic systemic inflammation in uremia: potential therapeutic approaches. Semin Nephrol 2004; 24:441.
  144. Kovesdy CP, Kalantar-Zadeh K. Novel targets and new potential: developments in the treatment of inflammation in chronic kidney disease. Expert Opin Investig Drugs 2008; 17:451.
  145. Vernaglione L, Cristofano C, Muscogiuri P, Chimienti S. Does atorvastatin influence serum C-reactive protein levels in patients on long-term hemodialysis? Am J Kidney Dis 2004; 43:471.
  146. Seliger SL, Weiss NS, Gillen DL, et al. HMG-CoA reductase inhibitors are associated with reduced mortality in ESRD patients. Kidney Int 2002; 61:297.
  147. Kumar S, Raftery M, Yaqoob M, Fan SL. Anti-inflammatory effects of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitors (statins) in peritoneal dialysis patients. Perit Dial Int 2007; 27:283.
  148. Deng J, Wu Q, Liao Y, et al. Effect of statins on chronic inflammation and nutrition status in renal dialysis patients: a systematic review and meta-analysis. Nephrology (Carlton) 2012; 17:545.
  149. Chang JW, Yang WS, Min WK, et al. Effects of simvastatin on high-sensitivity C-reactive protein and serum albumin in hemodialysis patients. Am J Kidney Dis 2002; 39:1213.
  150. Wanner C, Krane V, März W, et al. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med 2005; 353:238.
  151. Fellström BC, Jardine AG, Schmieder RE, et al. Rosuvastatin and cardiovascular events in patients undergoing hemodialysis. N Engl J Med 2009; 360:1395.
  152. Baigent C, Landry M. Study of Heart and Renal Protection (SHARP). Kidney Int Suppl 2003; :S207.
  153. Baigent C, Landray MJ, Reith C, 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.
  154. Storey BC, Staplin N, Haynes R, et al. Lowering LDL cholesterol reduces cardiovascular risk independently of presence of inflammation. Kidney Int 2018; 93:1000.
  155. Patel V, Joharapurkar A, Jain M. Role of mineralocorticoid receptor antagonists in kidney diseases. Drug Dev Res 2021; 82:341.
  156. Stenvinkel P, Andersson P, Wang T, et al. Do ACE-inhibitors suppress tumour necrosis factor-alpha production in advanced chronic renal failure? J Intern Med 1999; 246:503.
  157. Agarwal R. Proinflammatory effects of oxidative stress in chronic kidney disease: role of additional angiotensin II blockade. Am J Physiol Renal Physiol 2003; 284:F863.
  158. Bakris GL, Agarwal R, Anker SD, et al. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N Engl J Med 2020; 383:2219.
  159. Pitt B, Filippatos G, Agarwal R, et al. Cardiovascular Events with Finerenone in Kidney Disease and Type 2 Diabetes. N Engl J Med 2021; 385:2252.
  160. Epstein M, Kovesdy CP, Clase CM, et al. Aldosterone, Mineralocorticoid Receptor Activation, and CKD: A Review of Evolving Treatment Paradigms. Am J Kidney Dis 2022; 80:658.
  161. Alicic RZ, Neumiller JJ, Johnson EJ, et al. Sodium-Glucose Cotransporter 2 Inhibition and Diabetic Kidney Disease. Diabetes 2019; 68:248.
  162. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. Dapagliflozin in Patients with Chronic Kidney Disease. N Engl J Med 2020; 383:1436.
  163. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N Engl J Med 2019; 381:1995.
  164. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med 2017; 377:644.
  165. Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med 2019; 380:2295.
  166. Wanner C, Inzucchi SE, Lachin JM, et al. Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. N Engl J Med 2016; 375:323.
  167. Wiviott SD, Raz I, Bonaca MP, et al. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2019; 380:347.
  168. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med 2015; 373:2117.
  169. The EMPA-KIDNEY Collaborative Group, Herrington WG, Staplin N, et al. Empagliflozin in Patients with Chronic Kidney Disease. N Engl J Med 2023; 388:117.
  170. Bonnet F, Scheen AJ. Effects of SGLT2 inhibitors on systemic and tissue low-grade inflammation: The potential contribution to diabetes complications and cardiovascular disease. Diabetes Metab 2018; 44:457.
  171. Bonora BM, Avogaro A, Fadini GP. Extraglycemic Effects of SGLT2 Inhibitors: A Review of the Evidence. Diabetes Metab Syndr Obes 2020; 13:161.
  172. Dekkers CCJ, Petrykiv S, Laverman GD, et al. Effects of the SGLT-2 inhibitor dapagliflozin on glomerular and tubular injury markers. Diabetes Obes Metab 2018; 20:1988.
  173. Heerspink HJL, Perco P, Mulder S, et al. 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.
  174. Zoccali C, Mallamaci F. Novel Therapeutic Options for Cardiovascular Disease with CKD. Clin J Am Soc Nephrol 2021; 16:682.
  175. Ray KK, Nicholls SJ, Buhr KA, et al. Effect of Apabetalone Added to Standard Therapy on Major Adverse Cardiovascular Events in Patients With Recent Acute Coronary Syndrome and Type 2 Diabetes: A Randomized Clinical Trial. JAMA 2020; 323:1565.
  176. Kalantar-Zadeh K, Schwartz GG, Nicholls SJ, et al. Effect of Apabetalone on Cardiovascular Events in Diabetes, CKD, and Recent Acute Coronary Syndrome: Results from the BETonMACE Randomized Controlled Trial. Clin J Am Soc Nephrol 2021; 16:705.
  177. Boaz M, Smetana S, Weinstein T, et al. Secondary prevention with antioxidants of cardiovascular disease in endstage renal disease (SPACE): randomised placebo-controlled trial. Lancet 2000; 356:1213.
  178. Yang SK, Xiao L, Xu B, et al. Effects of vitamin E-coated dialyzer on oxidative stress and inflammation status in hemodialysis patients: a systematic review and meta-analysis. Ren Fail 2014; 36:722.
  179. Lines SW, Carter AM, Dunn EJ, et al. A randomized controlled trial evaluating the erythropoiesis stimulating agent sparing potential of a vitamin E-bonded polysulfone dialysis membrane. Nephrol Dial Transplant 2014; 29:649.
  180. Yang CC, Hsu SP, Wu MS, et al. Effects of vitamin C infusion and vitamin E-coated membrane on hemodialysis-induced oxidative stress. Kidney Int 2006; 69:706.
  181. Tepel M, van der Giet M, Statz M, et al. The antioxidant acetylcysteine reduces cardiovascular events in patients with end-stage renal failure: a randomized, controlled trial. Circulation 2003; 107:992.
  182. Saddadi F, Alatab S, Pasha F, et al. The effect of treatment with N-acetylcysteine on the serum levels of C-reactive protein and interleukin-6 in patients on hemodialysis. Saudi J Kidney Dis Transpl 2014; 25:66.
  183. Cepaityte D, Leivaditis K, Varouktsi G, et al. N-Acetylcysteine: more than preventing contrast-induced nephropathy in uremic patients-focus on the antioxidant and anti-inflammatory properties. Int Urol Nephrol 2023; 55:1481.
  184. Stockler-Pinto MB, Mafra D, Moraes C, et al. Brazil nut (Bertholletia excelsa, H.B.K.) improves oxidative stress and inflammation biomarkers in hemodialysis patients. Biol Trace Elem Res 2014; 158:105.
  185. Cardozo LF, Stockler-Pinto MB, Mafra D. Brazil nut consumption modulates Nrf2 expression in hemodialysis patients: A pilot study. Mol Nutr Food Res 2016; 60:1719.
  186. Moreira LSG, Brum ISDC, de Vargas Reis DCM, et al. Cinnamon: an aromatic condiment applicable to chronic kidney disease. Kidney Res Clin Pract 2023; 42:4.
  187. Moreira LSG, Fanton S, Cardozo L, et al. Pink pressure: beetroot (Beta vulgaris rubra) as a possible novel medical therapy for chronic kidney disease. Nutr Rev 2022; 80:1041.
  188. Alvarenga L, Salarolli R, Cardozo LFMF, et al. Impact of curcumin supplementation on expression of inflammatory transcription factors in hemodialysis patients: A pilot randomized, double-blind, controlled study. Clin Nutr 2020; 39:3594.
  189. Chermut TR, Fonseca L, Figueiredo N, et al. Effects of propolis on inflammation markers in patients undergoing hemodialysis: A randomized, double-blind controlled clinical trial. Complement Ther Clin Pract 2023; 51:101732.
  190. Baptista BG, Fanton S, Ribeiro M, et al. The effect of Brazilian Green Propolis extract on inflammation in patients with chronic kidney disease on peritoneal dialysis: A randomised double-blind controlled clinical trial. Phytomedicine 2023; 114:154731.
  191. Ribeiro M, Fanton S, Paiva BR, et al. Dark chocolate (70% cocoa) attenuates the inflammatory marker TNF-α in patients on hemodialysis. Clin Nutr ESPEN 2023; 53:189.
  192. Esgalhado M, Kemp JA, Paiva BR, et al. Resistant starch type-2 enriched cookies modulate uremic toxins and inflammation in hemodialysis patients: a randomized, double-blind, crossover and placebo-controlled trial. Food Funct 2020; 11:2617.
  193. Jun M, Venkataraman V, Razavian M, et al. Antioxidants for chronic kidney disease. Cochrane Database Syst Rev 2012; 10:CD008176.
  194. Ferramosca E, Burke S, Chasan-Taber S, et al. Potential antiatherogenic and anti-inflammatory properties of sevelamer in maintenance hemodialysis patients. Am Heart J 2005; 149:820.
  195. Yamada K, Fujimoto S, Tokura T, et al. Effect of sevelamer on dyslipidemia and chronic inflammation in maintenance hemodialysis patients. Ren Fail 2005; 27:361.
  196. Vlassara H, Uribarri J, Cai W, et al. Effects of sevelamer on HbA1c, inflammation, and advanced glycation end products in diabetic kidney disease. Clin J Am Soc Nephrol 2012; 7:934.
  197. Block GA, Spiegel DM, Ehrlich J, et al. Effects of sevelamer and calcium on coronary artery calcification in patients new to hemodialysis. Kidney Int 2005; 68:1815.
  198. Basutkar RS, Varghese R, Mathew NK, et al. Systematic review and meta-analysis of potential pleiotropic effects of sevelamer in chronic kidney disease: Beyond phosphate control. Nephrology (Carlton) 2022; 27:337.
  199. Tyrrell DJ, Horne AP, Holme KR, et al. Heparin in inflammation: potential therapeutic applications beyond anticoagulation. Adv Pharmacol 1999; 46:151.
  200. Lever R, Hoult JR, Page CP. The effects of heparin and related molecules upon the adhesion of human polymorphonuclear leucocytes to vascular endothelium in vitro. Br J Pharmacol 2000; 129:533.
  201. Bossola M, Muscaritoli M, Tazza L, et al. Malnutrition in hemodialysis patients: what therapy? Am J Kidney Dis 2005; 46:371.
  202. Lambert CP, Sullivan DH, Evans WJ. Effects of testosterone replacement and/or resistance training on interleukin-6, tumor necrosis factor alpha, and leptin in elderly men ingesting megestrol acetate: a randomized controlled trial. J Gerontol A Biol Sci Med Sci 2003; 58:165.
  203. Mantovani G, Macciò A, Lai P, et al. Cytokine involvement in cancer anorexia/cachexia: role of megestrol acetate and medroxyprogesterone acetate on cytokine downregulation and improvement of clinical symptoms. Crit Rev Oncog 1998; 9:99.
  204. Mantovani G. Does megestrol acetate down-regulate interleukin-6 in patients? Support Care Cancer 2002; 10:566.
  205. Yeh SS, Wu SY, Levine DM, et al. The correlation of cytokine levels with body weight after megestrol acetate treatment in geriatric patients. J Gerontol A Biol Sci Med Sci 2001; 56:M48.
  206. Boccanfuso JA, Hutton M, McAllister B. The effects of megestrol acetate on nutritional parameters in a dialysis population. J Ren Nutr 2000; 10:36.
  207. Burrowes JD, Bluestone PA, Wang J, Pierson RN Jr. The effects of moderate doses of megestrol acetate on nutritional status and body composition in a hemodialysis patient. J Ren Nutr 1999; 9:89.
  208. Rammohan M, Kalantar-Zadeh K, Liang A, Ghossein C. Megestrol acetate in a moderate dose for the treatment of malnutrition-inflammation complex in maintenance dialysis patients. J Ren Nutr 2005; 15:345.
  209. Gołębiewska JE, Lichodziejewska-Niemierko M, Aleksandrowicz-Wrona E, et al. Influence of megestrol acetate on nutrition, inflammation and quality of life in dialysis patients. Int Urol Nephrol 2012; 44:1211.
  210. Pergola PE, Raskin P, Toto RD, et al. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N Engl J Med 2011; 365:327.
  211. de Zeeuw D, Akizawa T, Audhya P, et al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 2013; 369:2492.
  212. Warady BA, Pergola PE, Agarwal R, et al. Effects of Bardoxolone Methyl in Alport Syndrome. Clin J Am Soc Nephrol 2022; 17:1763.
  213. Noori N, Dukkipati R, Kovesdy CP, et al. Dietary omega-3 fatty acid, ratio of omega-6 to omega-3 intake, inflammation, and survival in long-term hemodialysis patients. Am J Kidney Dis 2011; 58:248.
  214. Hu C, Yang M, Zhu X, et al. Effects of Omega-3 Fatty Acids on Markers of Inflammation in Patients With Chronic Kidney Disease: A Controversial Issue. Ther Apher Dial 2018; 22:124.
  215. Buoite Stella A, Gortan Cappellari G, Barazzoni R, Zanetti M. Update on the Impact of Omega 3 Fatty Acids on Inflammation, Insulin Resistance and Sarcopenia: A Review. Int J Mol Sci 2018; 19.
  216. Miroult C, Lasserre J, Toma S. Effects of Omega-3 as an adjuvant in the treatment of periodontal disease: A systematic review and meta-analysis. Clin Exp Dent Res 2023; 9:545.
  217. Kovesdy CP, Kalantar-Zadeh K. Vitamin D receptor activation and survival in chronic kidney disease. Kidney Int 2008; 73:1355.
  218. Donate-Correa J, Domínguez-Pimentel V, Méndez-Pérez ML, et al. Selective vitamin D receptor activation as anti-inflammatory target in chronic kidney disease. Mediators Inflamm 2014; 2014:670475.
  219. Alvarez JA, Zughaier SM, Law J, et al. Effects of high-dose cholecalciferol on serum markers of inflammation and immunity in patients with early chronic kidney disease. Eur J Clin Nutr 2013; 67:264.
  220. Miskulin DC, Majchrzak K, Tighiouart H, et al. Ergocalciferol Supplementation in Hemodialysis Patients With Vitamin D Deficiency: A Randomized Clinical Trial. J Am Soc Nephrol 2016; 27:1801.
  221. Zhao L, Zhu G, Wu L, Xie D. Effects of vitamin D on inflammatory state in patients with chronic kidney disease: A controversial issue. Ther Apher Dial 2023; 27:383.
  222. Chen Y, Abbate M, Tang L, et al. L-Carnitine supplementation for adults with end-stage kidney disease requiring maintenance hemodialysis: a systematic review and meta-analysis. Am J Clin Nutr 2014; 99:408.
  223. Hamedi-Kalajahi F, Imani H, Mojtahedi S, Shabbidar S. Effect of L-Carnitine Supplementation on Inflammatory Markers and Serum Glucose in Hemodialysis Children: A Randomized, Placebo-Controlled Clinical Trial. J Ren Nutr 2022; 32:144.
  224. Mosterd CM, Kanbay M, van den Born BJH, et al. Intestinal microbiota and diabetic kidney diseases: the Role of microbiota and derived metabolites inmodulation of renal inflammation and disease progression. Best Pract Res Clin Endocrinol Metab 2021; 35:101484.
  225. Sumida K, Lau WL, Kovesdy CP, et al. Microbiome modulation as a novel therapeutic approach in chronic kidney disease. Curr Opin Nephrol Hypertens 2021; 30:75.
  226. Tian N, Li L, Ng JK, Li PK. The Potential Benefits and Controversies of Probiotics Use in Patients at Different Stages of Chronic Kidney Disease. Nutrients 2022; 14.
  227. Favero C, Ortiz A, Sanchez-Niño MD. Probiotics for kidney disease. Clin Kidney J 2022; 15:1981.
  228. Lopes RCSO, Balbino KP, Jorge MP, et al. Modulation of intestinal microbiota, control of nitrogen products and inflammation by pre/probiotics in chronic kidney disease: a systematic review. Nutr Hosp 2018; 35:722.
  229. Thongprayoon C, Kaewput W, Hatch ST, et al. Effects of Probiotics on Inflammation and Uremic Toxins Among Patients on Dialysis: A Systematic Review and Meta-Analysis. Dig Dis Sci 2019; 64:469.
  230. Mafra D, Borges NA, Lindholm B, et al. Food as medicine: targeting the uraemic phenotype in chronic kidney disease. Nat Rev Nephrol 2021; 17:153.
  231. Alvarenga L, Cardozo LFMF, Borges NA, et al. Can nutritional interventions modulate the activation of the NLRP3 inflammasome in chronic kidney disease? Food Res Int 2020; 136:109306.
  232. He LX, Abdolmaleky HM, Yin S, et al. Dietary Fermented Soy Extract and Oligo-Lactic Acid Alleviate Chronic Kidney Disease in Mice via Inhibition of Inflammation and Modulation of Gut Microbiota. Nutrients 2020; 12.
  233. Joshi S, Moore LW, Kalantar-Zadeh K. The Future of Nutrition in Kidney Disease: Plant-Based Diets, Gut Microbiome, and Beyond. J Ren Nutr 2021; 31:97.
  234. Joshi S, McMacken M, Kalantar-Zadeh K. Plant-Based Diets for Kidney Disease: A Guide for Clinicians. Am J Kidney Dis 2021; 77:287.
  235. Don BR, Kim K, Li J, et al. The effect of etanercept on suppression of the systemic inflammatory response in chronic hemodialysis patients. Clin Nephrol 2010; 73:431.
  236. Identifier: NCT 00293202 www.clinicaltrials.gov (Accessed on June 02, 2008).
  237. Kiely PD, Gillespie KM, Oliveira DB. Oxpentifylline inhibits tumor necrosis factor-alpha mRNA transcription and protects against arthritis in mercuric chloride-treated brown Norway rats. Eur J Immunol 1995; 25:2899.
  238. Rattanasompattikul M, Molnar MZ, Lee ML, et al. Anti-Inflammatory and Anti-Oxidative Nutrition in Hypoalbuminemic Dialysis Patients (AIONID) study: results of the pilot-feasibility, double-blind, randomized, placebo-controlled trial. J Cachexia Sarcopenia Muscle 2013; 4:247.
  239. Antunes SA, Vilela RQ, Vaz JD, Canziani ME. Pentoxifylline does not alter the concentration of hepcidin in chronic kidney disease patients undergoing hemodialysis. Int J Artif Organs 2014; 37:521.
  240. Sabounjian L, Graham P, Wu L, et al. A First-in-Patient, Multicenter, Double-Blind, 2-Arm, Placebo-Controlled, Randomized Safety and Tolerability Study of a Novel Oral Drug Candidate, CTP-499, in Chronic Kidney Disease. Clin Pharmacol Drug Dev 2016; 5:314.
  241. Braman V, Graham P, Cheng C, et al. A Randomized Phase I Evaluation of CTP-499, a Novel Deuterium-Containing Drug Candidate for Diabetic Nephropathy. Clin Pharmacol Drug Dev 2013; 2:53.
  242. Yang BB, Baughman S, Sullivan JT. Pharmacokinetics of anakinra in subjects with different levels of renal function. Clin Pharmacol Ther 2003; 74:85.
  243. Hung AM, Ellis CD, Shintani A, et al. IL-1β receptor antagonist reduces inflammation in hemodialysis patients. J Am Soc Nephrol 2011; 22:437.
  244. Nowak KL, Chonchol M, Ikizler TA, et al. IL-1 Inhibition and Vascular Function in CKD. J Am Soc Nephrol 2017; 28:971.
  245. Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med 2017; 377:1119.
  246. Ridker PM, MacFadyen JG, Glynn RJ, et al. Inhibition of Interleukin-1β by Canakinumab and Cardiovascular Outcomes in Patients With Chronic Kidney Disease. J Am Coll Cardiol 2018; 71:2405.
  247. Ridker PM, Devalaraja M, Baeres FMM, 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.
  248. Pergola PE, Devalaraja M, Fishbane S, et al. Ziltivekimab for Treatment of Anemia of Inflammation in Patients on Hemodialysis: Results from a Phase 1/2 Multicenter, Randomized, Double-Blind, Placebo-Controlled Trial. J Am Soc Nephrol 2021; 32:211.
  249. Wada Y, Jensen C, Meyer ASP, et al. Efficacy and safety of interleukin-6 inhibition with ziltivekimab in patients at high risk of atherosclerotic events in Japan (RESCUE-2): A randomized, double-blind, placebo-controlled, phase 2 trial. J Cardiol 2023; 82:279.
  250. Cragg MS, Walshe CA, Ivanov AO, Glennie MJ. The biology of CD20 and its potential as a target for mAb therapy. Curr Dir Autoimmun 2005; 8:140.
  251. Cambridge G, Leandro MJ, Edwards JC, et al. Serologic changes following B lymphocyte depletion therapy for rheumatoid arthritis. Arthritis Rheum 2003; 48:2146.
  252. Edwards JC, Szczepanski L, Szechinski J, et al. Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med 2004; 350:2572.
  253. Cambridge G, Leandro MJ, Teodorescu M, et al. B cell depletion therapy in systemic lupus erythematosus: effect on autoantibody and antimicrobial antibody profiles. Arthritis Rheum 2006; 54:3612.
  254. Keogh KA, Wylam ME, Stone JH, Specks U. Induction of remission by B lymphocyte depletion in eleven patients with refractory antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum 2005; 52:262.
  255. Keogh KA, Ytterberg SR, Fervenza FC, et al. Rituximab for refractory Wegener's granulomatosis: report of a prospective, open-label pilot trial. Am J Respir Crit Care Med 2006; 173:180.
  256. Smith KG, Jones RB, Burns SM, Jayne DR. Long-term comparison of rituximab treatment for refractory systemic lupus erythematosus and vasculitis: Remission, relapse, and re-treatment. Arthritis Rheum 2006; 54:2970.
  257. Kamar N, Faguer S, Esposito L, et al. Treatment of focal segmental glomerular sclerosis with rituximab: 2 case reports. Clin Nephrol 2007; 67:250.
  258. Milovanova SIu, Lopatkina TN, Kozlovskaia LV, Krasnova TN. [Monoclonal antibodies to B-lymphocytes (rituximab) in the treatment of HCV-associated severe cryoglobulinemic glomerulonephritis]. Ter Arkh 2007; 79:69.
  259. Ponticelli C. New therapies for lupus nephritis. Clin J Am Soc Nephrol 2006; 1:863.
  260. Ruggenenti P, Chiurchiu C, Abbate M, et al. Rituximab for idiopathic membranous nephropathy: who can benefit? Clin J Am Soc Nephrol 2006; 1:738.
  261. Smith RJ, Alexander J, Barlow PN, et al. New approaches to the treatment of dense deposit disease. J Am Soc Nephrol 2007; 18:2447.
  262. de Zeeuw D, Bekker P, Henkel E, et al. The effect of CCR2 inhibitor CCX140-B on residual albuminuria in patients with type 2 diabetes and nephropathy: a randomised trial. Lancet Diabetes Endocrinol 2015; 3:687.
  263. Menne J, Eulberg D, Beyer D, et al. C-C motif-ligand 2 inhibition with emapticap pegol (NOX-E36) in type 2 diabetic patients with albuminuria. Nephrol Dial Transplant 2017; 32:307.
  264. Perez-Gomez MV, Sanchez-Niño MD, Sanz AB, et al. Targeting inflammation in diabetic kidney disease: early clinical trials. Expert Opin Investig Drugs 2016; 25:1045.
  265. Shinozaki Y, Fukui K, Kobayashi H, et al. JTZ-951 (enarodustat), a hypoxia-inducible factor prolyl hydroxylase inhibitor, improves iron utilization and anemia of inflammation: Comparative study against recombinant erythropoietin in rat. Eur J Pharmacol 2021; 898:173990.
  266. Yan Z, Xu G. A Novel Choice to Correct Inflammation-Induced Anemia in CKD: Oral Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor Roxadustat. Front Med (Lausanne) 2020; 7:393.
  267. Joharapurkar AA, Patel VJ, Kshirsagar SG, et al. Prolyl hydroxylase inhibitor desidustat protects against acute and chronic kidney injury by reducing inflammatory cytokines and oxidative stress. Drug Dev Res 2021; 82:852.
  268. Bloembergen WE, Hakim RM, Stannard DC, et al. Relationship of dialysis membrane and cause-specific mortality. Am J Kidney Dis 1999; 33:1.
  269. Maduell F, Moreso F, Pons M, et al. High-efficiency postdilution online hemodiafiltration reduces all-cause mortality in hemodialysis patients. J Am Soc Nephrol 2013; 24:487.
  270. Castaneda C, Gordon PL, Parker RC, et al. Resistance training to reduce the malnutrition-inflammation complex syndrome of chronic kidney disease. Am J Kidney Dis 2004; 43:607.
  271. Barcellos FC, Del Vecchio FB, Reges A, et al. Exercise in patients with hypertension and chronic kidney disease: a randomized controlled trial. J Hum Hypertens 2018; 32:397.
  272. Gadelha AB, Cesari M, Corrêa HL, et al. Effects of pre-dialysis resistance training on sarcopenia, inflammatory profile, and anemia biomarkers in older community-dwelling patients with chronic kidney disease: a randomized controlled trial. Int Urol Nephrol 2021; 53:2137.
  273. Rosa TS, Corrêa HL, Deus LA, et al. Effects of dynamic and isometric resistance training protocols on metabolic profile in hemodialysis patients: a randomized controlled trial. Appl Physiol Nutr Metab 2021; 46:1029.
  274. de Araújo TB, de Luca Corrêa H, de Deus LA, et al. The effects of home-based progressive resistance training in chronic kidney disease patients. Exp Gerontol 2023; 171:112030.
Topic 1939 Version 29.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟