Abstract

Elevated plasma levels of the phosphaturic hormone fibroblast growth factor 23 (FGF-23) are a hallmark of chronic kidney disease (CKD)-mineral and bone disorder. FGF-23 allows serum phosphate levels within physiological limits to be maintained in progressive CKD until end-stage renal disease is reached. Despite its seemingly beneficial role in phosphate homeostasis, several prospective studies in dialysis patients and in patients with less advanced CKD associated elevated FGF-23 with poor cardiovascular and renal outcome. Moreover, very recent evidence suggests an adverse prognostic impact of elevated FGF-23 even in subjects without manifest CKD. These epidemiological data are supplemented by laboratory findings that reveal a pathophysiological role of FGF-23 in the pathogenesis of myocardial injury. In aggregate, these clinical and experimental data identify FGF-23 as a promising target of novel therapeutic interventions in CKD and beyond, which should be tested in future clinical trials.

Introduction

Patients with chronic kidney disease (CKD) suffer from tremendous cardiovascular morbidity and mortality. This is exemplarily illustrated by recent data from the ERA-EDTA Registry which found 35-year-old incident dialysis patients to share the cardiovascular prognosis with 75-year-old subjects from the general population [1]. Numerous clinical cohort studies published in the last decade indicate that cardiovascular morbidity may even increase in much earlier CKD stages, as summarized by the Chronic Kidney Disease Prognosis Consortium [2].

It is generally acknowledged that this high cardiovascular risk in CKD patients is not merely caused by ‘traditional’ risk factors that drive vascular disease in the general population—namely smoking, hyperglycaemia, hypercholesterolaemia, arterial hypertension, obesity and physical inactivity [3]. In line with this assumption, prediction models derived from the general population do not adequately predict future cardiovascular events among CKD patients [4]. Instead, CKD-specific risk factors undoubtedly contribute to cardiovascular morbidity in these patients [3].

This concept is corroborated by the failure of most interventional studies which aimed to target traditional cardiovascular risk factors in CKD [5–7]. Thus, a clinical update from Kidney Disease: Improving Global Outcome advocated to clarify the role of novel risk factors as potential new therapeutic targets [3].

A considerable number of CKD-specific risk factors have been defined in recent years. Unfortunately, some of these factors are hard to tackle therapeutically—such as chronic microinflammation—while others have been targets of recent trials which uniformly yielded disappointing results: correction of anaemia [8], vitamin substitution in hyperhomocysteinaemia [9–11], early dialysis initiation [12], intensification of dialysis treatment or the use of modern dialysis equipment [13] failed to improve prognosis of CKD patients.

Against this background, CKD-mineral and bone disorder (CKD-MBD) represent a welcome exception: first, several components of CKD-MBD have consistently been associated with adverse outcome in epidemiological studies [14–17]. Secondly, therapeutic agents for CKD-MBD are widely available. Thirdly, cohort studies [18] and preliminary interventional trials on surrogate markers [19–21] suggest that modification of the CKD-MBD parameters may be beneficial, although definite data from adequately powered studies analysing ‘hard’ cardiovascular endpoints are pending.

Our conceptual understanding of CKD-MBD has been challenged by the discovery of the phosphaturic hormone fibroblast growth factor 23 (FGF-23). Only 12 years after its first description, most experts agree that FGF-23 plays a central role in CKD-MBD, and very recent data suggest a direct pathophysiological link between FGF-23 and cardiovascular disease in CKD.

In the present paper, we will first give a short summary of the physiological role of FGF-23 in human calcium–phosphate metabolism. Several detailed reviews on this topic have been published in the last years, to which the interested reader may refer [22–25]. Next, we will discuss the tremendous rise in FGF-23 levels (for which the term ‘hyperphosphatoninism’ was coined) in CKD patients, and we will review epidemiological studies which associated hyperphosphatoninism with prevalent and incident cardiovascular disease. Finally, we will summarize models which offer pathophysiological explanations for these associations, and discuss FGF-23 as a future therapeutic target in CKD.

Physiological functions of FGF-23

FGF-23 is a 251-amino acid protein with a molecular weight of 26 000 Da, which is mainly synthesized and secreted by osteoblasts [26]. Together with FGF-19 and FGF-21, FGF-23 belongs to the FGF-19 subfamily of FGFs. Unlike most other FGFs, which bind to heparin sulphate in the extracellular matrix and thus exert paracrine and autocrine effects, members of the FGF-19 subfamily display topological differences in the heparin-binding region. Consequently, FGF-23 is less avidly captured in the extracellular matrix, and thus more prone to enter the systemic circulation, allowing FGF-23 to exert endocrine functions [27, 28].

It is generally assumed that hyperphosphataemia and/or oral phosphate loading induces osteoblastic FGF-23 secretion, although the exact mechanisms remain enigmatic at present. Calcitriol and parathyroid hormone (PTH) are additional regulators of FGF-23 secretion, as discussed below.

FGF-23 exerts its physiological functions by binding to FGF receptors (FGF-R) on the surface of target cells in the presence of membrane-bound Klotho, which serves as a co-receptor. Of note, FGF-Rs are found ubiquitously. In contrast, Klotho is selectively expressed in kidneys, parathyroid glands and in the choroid plexus [29], rendering these organs physiological targets for FGF-23.

In the kidney, FGF-23 mediates its phosphaturic effect by reducing the expression and activity of sodium–phosphate (Na/Pi) cotransporters in the proximal tubules [30]. Additionally, FGF-23 may reduce intestinal NaPi cotransporter activity and thus directly inhibit intestinal phosphate absorption [31].

Furthermore, FGF-23 lowers serum calcitriol levels by decreasing the renal expression of 1α-hydroxylase (CYP27B1), which is the rate-limiting step in calcitriol synthesis, and increasing the expression of 24-hydroxylase (CYP24A1), which degrades calcitriol [32]. As calcitriol itself stimulates FGF-23 generation by binding to the FGF-23 gene promoter [33], a regulatory feedback loop between FGF-23 and calcitriol exists.

Finally, FGF-23 suppresses PTH synthesis in vitro [34] and in vivo [35]. However, parathyroid glands may become resistant towards FGF-23 in uraemia, as PTH levels are elevated in the presence of severe hyperphosphatoninism in uraemic rodent models [36].

In contrast, it is uncertain whether PTH directly affects FGF-23 secretion: no effect of PTH on FGF-23 expression in bone cells was found in vitro [33, 37, 38] and conflicting in vivo data on the impact of PTH on FGF-23 levels have been reported from animal studies [37, 39] and in clinical trials [40–42]. Physiological interactions may partly explain these contradictory data, as PTH directly modifies phosphate and calcitriol levels, which themselves affect FGF-23 secretion. It has been suggested that a biphasic effect may exist, with an initial decrease in FGF-23 levels after PTH application to counteract PTH-induced phosphaturia, and a later increase in FGF-23 [41]. In summary, the exact regulatory circuit between FGF-23 and PTH is not fully understood yet. The central role of FGF-23 in phosphate metabolism is summarized in Figure 1.

Fig. 1.

Serum phosphate regulation. Black arrows indicate activating pathways; red lines indicate inhibitory pathway. Note the exact pathways by which phosphate triggers PTH and FGF-23 secretion, and the direct link between PTH and FGF-23 await further experimental studies, as indicated by dotted arrows. Details are discussed in the manuscript.

Measurement of FGF-23

Two assays for quantification of human FGF-23 are commercially available. Full-length (‘intact’) FGF-23 levels can be determined with a sandwich enzyme-linked immunosorbent assay, in which two different monoclonal antibodies detect the simultaneous presence of both the N-terminal and C-terminal portions of FGF-23. In contrast, the ‘C-terminal’ assay recognizes both full-length and processed C-terminal fragments of FGF-23. Measurement results from the ‘intact’ and the ‘C-terminal’ assay are strongly correlated, and most experts agree that the use of either of these assays is adequate. For the ease of legibility, the present review will not indicate the specific assay used in individual clinical studies.

FGF-23 in CKD

A progressive rise in FGF-23 occurs over the spectrum of CKD [43, 44], and maintenance dialysis patients may display FGF-23 levels which are more than 100-fold higher compared with healthy subjects [43]. Thereby, FGF-23 levels of most dialysis patients by far surpass levels that are observed in patients with hereditary diseases of phosphate metabolism, such as hypophosphataemic rickets [45].

Recent papers expanded these findings and reported an increment in FGF-23 levels already at very early stages of CKD [46]. More than half of all CRIC (Chronic Renal Insufficiency Cohort Study) participants with slightly reduced kidney function [defined as estimated glomerular filtration rate (eGFR) 50–59 mL/min] had elevated FGF-23 levels, while PTH was elevated only in a minority of these patients, and virtually, no patient had hyperphosphataemia [47]. Elevated FGF-23 is therefore considered as the earliest detectable change in CKD-MBD.

Still, it remains unclear which stimulus drives this increase in FGF-23 in the early course of CKD. Two opposing hypotheses were raised: either kidney injury itself may induce the production of a—yet to be defined—factor that stimulates FGF-23 section (or alternatively, decreased the production of a tonic inhibitor) [47]. This hypothesis is partly supported by animal data that demonstrate an early increase in FGF-23 after kidney injury [48]. Alternatively, increased levels of FGF-23 may reflect dampened Klotho expression in target organs [49, 50]. In line with this hypothesis, decreased renal Klotho expression has been detected in kidney biopsy studies [51]. As FGF-23 requires Klotho for its phosphate-regulating effects in kidneys and in parathyroid glands, higher FGF-23 might physiologically be needed to overcome Klotho deficiency and to maintain normophosphataemia in states of low Klotho expression.

If this second hypothesis held true, hyperphosphataemia and hypervitaminosis D should at least transiently occur in early CKD as a reflection of such initial Klotho deficiency [47]. Of note, large epidemiological data stand against this hypothesis, as low rather than high serum phosphate values are found in very early CKD [47]. As FGF-23 itself down-regulates Klotho expression [52], Klotho deficiency in early CKD might thus reflect a primary increase in FGF-23.

Implications of hyperphosphatoninism in CKD

Counterintuitive to the presumptively protective, phosphate-regulating properties of FGF-23, several longitudinal studies associated hyperphosphatoninism with adverse renal outcome and mortality in CKD patients.

First prospective data came from the Mild to Moderate Kidney Disease (MMKD) study, which investigated the relationship between FGF-23 and CKD progression in 177 non-diabetic CKD stage 1–5 patients not receiving dialysis therapy [53]. Over a median follow-up of 4.4 years, patients with elevated FGF-23 (defined as C-terminal FGF-23 >104 rU/mL and/or intact FGF-23 >35 pg/mL) more often reached the primary combined endpoint defined as doubling of serum creatinine and/or onset of renal replacement therapy. Given the strong association between FGF-23 and renal function at the baseline, such a correlation may be expected in univariate analysis. However, this association remained significant after adjustment for confounders including eGFR. Similar findings were reported in a smaller cohort of 55 diabetic CKD patients prospectively followed for 31 months [54].

Subsequently, the association between FGF-23 and renal outcome was re-analysed in two large longitudinal cohort studies, namely CRIC [55] (3879 CKD stage 2–4 patients; median follow-up 3.5 years) and HOST [56] (Homocysteine in Kidney and End-Stage Renal Disease study; 1099 CKD stage 4–5 patients not on dialysis; median follow-up 2.9 years), which yielded partly conflicting results. Both studies assessed renal outcome (defined as initiation of dialysis treatment or renal transplantation in CRIC, and as initiation of dialysis treatment in HOST) and patient survival; HOST additionally reported cardiovascular events. In line with MMKD, both studies found high FGF-23 as a predictor of adverse renal outcome in univariate analyses. This relationship lost significance after adjusting for eGFR in the CRIC study, but not in HOST. In subgroup analyses, elevated FGF-23 levels remained an independent renal predictor in CRIC participants with a baseline eGFR of >30 mL/min/1.73 m2, but not in patients with more severe CKD at the baseline [55].

Nonetheless, CRIC and HOST consistently found an increasing mortality risk at higher quartiles of FGF-23. Compared with the lowest FGF-23 quartile, patients in the fourth quartile suffered from a more than 2-fold higher mortality after adjustment for classical cardiovascular risk factors and traditional CKD-MBD markers [55, 56].

These findings are congruent with earlier data from cohort studies in CKD 5D patients: in a nested case–control analysis within the Accelerated Mortality on Renal Replacement (ArMORR) study, Gutierrez et al. [57] measured FGF-23 levels of 400 incident haemodialysis patients. Elevated FGF-23 levels not only independently predicted 1-year all-cause mortality, but even surpassed serum phosphate levels as a marker of adverse outcome [45, 57]. Similar findings were reported in a cohort of 219 prevalent dialysis patients followed for 2 years [58]; notably, a third dialysis study reported FGF-23 to predict survival only among male patients with prevalent cardiovascular disease [59].

FGF-23 and cardiovascular outcome

Unfortunately, none of these CKD studies that associated elevated FGF-23 with higher mortality reported specific causes of death [55–59], and only HOST prospectively analysed cardiovascular events [56]. Here, patients in the highest quartile of baseline FGF-23 had a hazard ratio of 2.58 for future cardiovascular events in univariate analysis, compared with patients with lowest FGF-23. In multivariate analysis, high FGF-23 remained a significant predictor of cardiovascular outcome, while native vitamin D, calcitriol and PTH did not. When separating the composite cardiovascular endpoint into its individual components, elevated FGF-23 predicted myocardial infarction and lower extremity amputation, but not thrombotic stroke [56].

These findings are in agreement with an earlier analysis from our group among 149 CKD stage 1–5 non-dialysis patients, in which elevated FGF-23 independently predicted a predefined combined cardiovascular endpoint. Our study was not powered to separately analyse mortality or individual components of the combined cardiovascular endpoint [60]. Finally, the Osaka Vitamin D Study in Patients with CKD (OVIDS-CKD) very recently aimed to extend these findings to Asian patients. OVIDS-CKD recruited 738 CKD stage 1–5 patients not on dialysis from Japan and again found high FGF-23 to predict broadly defined cardiovascular events when censoring patients at initiation of chronic dialysis treatment. This finding was less evident when further following patients after the onset of dialysis treatment, and analysis of cause-specific mortality was hampered by the low cardiovascular death toll [61].

In summary, across the stages of CKD, FGF-23 may predict all-cause mortality, cardiovascular event rate and renal progression. These findings inspired several groups to inquire whether a similar prognostic role of FGF-23 may exist in subjects without overt CKD. Such an idea follows recent reports which identified other components of CKD-MBD (e.g. serum phosphate [62–65] and hypovitaminosis D [66]) as predictors of adverse outcome in subjects without overt kidney disease.

Presently, longitudinal data from three cohort studies [namely the Heart and Soul Study [67], Health Professionals Follow-up Study (HPFS) [68] and the Women's Health and Aging Study (WHAS) [69]] have been published. Results from the Prevention of Events with Angiotensin Converting Enzyme Inhibition (PEACE) trial were presented at the American College of Cardiology's 61st Annual Scientific Session [70], and data from our HOM SWEET HOMe study will be submitted for presentation at the American Society of Nephrology's kidney week in late 2012.

In the Heart and Soul Study, FGF-23 was measured in 833 outpatients with prevalent coronary heart disease, who were followed for a median of 6 years. High FGF-23 at the baseline independently predicted adverse outcome. After adjustment for confounders, risk for incident cardiovascular events and mortality was doubled in the highest FGF-23 tertile compared with the lowest tertile [67]. A nested case–control analysis from HPFS yielded seemingly conflicting results: baseline FGF-23 levels did not differ between 422 study participants who subsequently suffered a coronary event and 837 age-matched controls who had no event over a follow-up period of 10 years [68].

Next, the WHAS study analysed FGF-23 in 701 moderately to severely disabled women (half of whom had impaired eGFR at baseline), who were followed for a median of 18 months. Among those women with eGFR ≥60 mL/min/1.73 m2 at the baseline, higher baseline FGF-23 was associated with incident CKD during follow-up. In contrast, FGF-23 did not independently predict global or cause-specific mortality. Of note, only 18.6% deaths were attributed to cardiovascular disease, which may result from selective inclusion of disabled women. Short follow-up, missing data on non-fatal cardiovascular events and high prevalence of CKD at the baseline are further limitations, which in aggregate preclude an uncritical transfer of these results from highly selected disabled seniors to subjects from the general population.

Instead, the discrepant results from the Heart and Soul Study [67] and from HPFS [68] warrant a critical review of major differences in study design. First, the Heart and Soul Study recruited patients with prevalent coronary artery disease, whereas HPFS included subjects without cardiovascular disease at the baseline. Secondly, different endpoints were assessed, as HPFS focused on coronary events (non-fatal acute myocardial infarction and fatal coronary heart disease), whereas Heart and Soul analysed a combined endpoint which comprised acute myocardial infarction and several non-coronary events (namely stroke, transient ischaemic attacks and admission for heart failure). Interestingly, the Heart and Soul Study investigators reported the impact of FGF-23 measurements on individual components of the combined endpoints separately; elevated FGF-23 selectively predicted non-coronary events, particularly admission for heart failure, while it failed to predict acute myocardial infarction—quite in line with HPFS. These findings are supplemented by very recently presented data from the PEACE trial [70]: PEACE randomized 8290 patients with stable coronary artery disease and normal or near-normal left ventricular function to intake of the ACE inhibitor trandolapril or placebo. The PEACE trial investigators now performed a nested case–control study which comprised 1100 out of 8290 PEACE participants, in whom FGF-23 and traditional cardiovascular biomarkers were analysed from samples drawn at study initiation. Preliminary results suggest that FGF-23 independently predicts cardiovascular death and incident heart failure, but not acute myocardial infarction or stroke.

In aggregate, these large prospective studies suggest that high FGF-23 levels may be associated with incident heart failure and cardiovascular death, rather than with incident coronary events, in individuals without overt CKD. As discussed below, these findings are in line with cross-sectional data from our HOM SWEET HOMe study, which is so far the largest non-CKD cohort in which the impact of FGF-23 on prevalent and incident cardiovascular disease is analysed [71].

FGF-23: marker or mediator of cardiovascular outcome?

The consistent association of hyperphosphatoninism with patient mortality across the spectrum of CKD, as well as with heart failure and (cardiovascular) mortality in several non-CKD patient cohorts, raises the question whether FGF-23 merely represents a ‘passive’ risk marker, or actively mediates cardiovascular damage.

In the former case, elevated FGF-23 levels might be an ‘innocent bystander’ that reflects the adverse cardiovascular implications of impaired phosphate metabolism and/or renal damage. Oral phosphate intake and hyperphosphataemia are considered major inducers of FGF-23 expression, and experimental data demonstrate that phosphate actively contributes to accelerated atherogenesis [72], arterial calcification [73] and left ventricular hypertrophy (LVH) [74]. Even though the prognostic impact of FGF-23 on cardiovascular events and/or mortality persisted after adjustment for serum phosphate in virtually all clinical trials, residual confounding cannot be ruled out: unlike FGF-23, serum phosphate measurements have a large biological variability [75]. Therefore, FGF-23 has been suggested to reflect serum phosphate levels over a longer period of time, similar to glycated haemoglobin in glucose homeostasis [76]. Thus, FGF-23 would be a more reliable indicator of elevated phosphate burden than a single phosphate measurement in epidemiological analyses.

In addition, as FGF-23 levels increase very early in the course of CKD, associations between elevated FGF-23 and adverse outcome in subjects without overt kidney disease might be caused by the well-known cardiovascular implications of even mild CKD. Creatinine-based estimations of GFR, which are used in most epidemiological studies for assessment of kidney function, do not always precisely detect early CKD. Moreover, all prospective studies that analysed hyperphosphatoninism in subjects without overt CKD did not specifically exclude subjects with eGFR <60 mL/min/1.73 m2. Despite adjustment for eGFR in multivariate analyses, residual confounding may occur because of limited precision of GFR estimation.

Alternatively, FGF-23 might actively induce cardiovascular damage, either via indirect or via direct pathways. Indirect pathways might imply hypovitaminosis D, as FGF-23 inhibits 1α-hydroxylase and thus lowers serum calcitriol levels, which is increasingly acknowledged as a non-traditional cardiovascular risk factor in subjects in CKD and non-CKD subjects.

The identification of direct pathophysiological pathways between hyperphosphatoninism and incident cardiovascular disease was more challenging, and our traditional understanding of FGF-23 biology seemingly precluded the existence of direct pathways: FGF-23 was deemed to require membrane-bound Klotho for exerting its endocrine effects at cellular level, and Klotho expression was considered to be restricted to traditional target organs of phosphate regulating hormones, namely to kidneys and parathyroid glands.

This traditional understanding was very recently refuted by Faul [77], who unveiled a Klotho-independent, causal role for FGF-23 in the pathogenesis of left ventricular hypertrophy (LVH).

Cardiovascular implications of hyperphosphatoninism

Inspired by cross-sectional echocardiographic studies which associated higher serum FGF-23 levels with prevalent LVH in CKD subjects and healthy subjects [78, 79], Faul et al. [80] set out to identify a direct cardiotoxic effect of FGF-23.

In a series of very elegant experiments, the authors first reported cell culture data which showed FGF-23 to directly cause pathological hypertrophic growth of isolated rat cardiomyocytes via FGF receptor-dependent activation of the calcineurin-NFAT (Nuclear factor of activated T-cells) signalling pathway, although cardiomyocytes did not express Klotho. Secondly, the authors replicated this pro-hypertrophic effect of FGF-23 on myocardial cells in vivo: in rodent models, intramyocardial as well as intravenous injection of FGF-23 induced pathological LVH. Similarly, LVH developed in Klotho-deficient mice, which constitutively overexpress FGF-23 in a futile attempt to combat hyperphosphataemia and hypervitaminosis D (which arise from Klotho deficiency and subsequent renal resistance to FGF-23 stimulation). Thirdly, the authors underscore the therapeutic potential of their findings in a rodent model of CKD, as administration of an unselective FGF-receptor blocker allowed the amelioration of the development of LVH in rats after 5/6 nephrectomy.

Finally, the experimental work was supplemented with a large set of serial echocardiographic studies from CRIC study participants. Expanding earlier findings from cross-sectional studies [78, 79], hyperphosphatoninism was associated with future LVH development in CKD patients who had normal ventricular geometry at study initiation.

LVH predisposes patients to subsequent development of left ventricular dysfunction and congestive heart failure [81, 82]. While experimental data on a direct pathophysiological contribution of hyperphosphatoninism in the sequential development of congestive heart failure are eagerly awaited, first large-scale clinical evidence for an association between elevated FGF-23 and impaired left ventricular systolic function came from our prospective HOM SWEET HOMe study.

In this study, we measured FGF-23 levels in 1309 patients who underwent elective coronary angiography between May 2007 and June 2010. Left ventricular function was assessed with ventriculography in 1022 out of 1309 patients. Cross-sectional data from all patients recruited before January 2010 were published recently [71] and up-dated for the present review by adding data from patients recruited between February and June 2010. In line with our initial report, HOM SWEET HOMe patients with impaired left ventricular function had higher FGF-23 levels; the same held true after exclusion of patients with an eGFR of <60 mL/min/1.73 m2 [71]. Notably, these findings are in line with supplementary information reported from the Heart and Soul Study, in which baseline ejection fraction was similarly correlated with serum FGF-23 levels.

Fig. 2.

FGF-23 levels among 1309 HOM SWEET HOMe participants after stratifying for ejection fraction (EF; categorized as 40, 40–59 and ≥60%) in the total study cohort (left columns), and in individuals with intact renal function (eGFR ≥60 mL/min/1.73 m2; right columns). Data are presented as mean + SEM.

We additionally re-analysed the association of hyperphosphatoninism with atrial fibrillation among all HOM SWEET HOMe participants. In line with our earlier report, higher FGF-23 levels were found in patients with prevalent atrial fibrillation before and after exclusion of patients with an eGFR of <60 mL/min/1.73 m2, again pointing towards a relevant conjunction between hyperphosphatoninism and cardiac damage (Figure 3).

Fig. 3.

FGF-23 levels among 1309 HOM SWEET HOMe participants after stratifying for the presence of atrial fibrillation (AF) in the total study cohort (left columns), and in individuals with intact renal function (eGFR ≥60 mL/min/1.73 m2; right columns). Data are presented as mean + SEM.

Notably, HOM SWEET HOMe did not find a correlation between serum FGF-23 levels and presence or severity of coronary artery disease. At a first glance, this finding may appear somewhat counterintuitive, given the strong association between FGF-23 and serum phosphate levels, and the large experimental and clinical evidence for a significant contribution of phosphate to cardiovascular disease [83]. Nevertheless, these findings are in line with cross-sectional studies in patients without overt CKD, which did not find a rise in serum FGF-23 levels among patients with coronary artery stenosis at coronary multislice computed tomography [84] or with the presence of arterial stenosis at whole-body magnetic resonance angiography [85] after adjustment for confounders.

Will FGF-23 become a novel therapeutic target in cardiovascular medicine?

Integrating recent pivotal experimental data on a Klotho-independent cardiotoxic impact of elevated FGF-23 into topical epidemiological evidence for a detrimental effect of hyperphosphatoninism, the following pathophysiological concept might be hypothesized: diet and/or CKD-associated chronic phosphate loading stimulates osteoblastic FGF-23 secretion. The resulting hyperphosphatoninism, which physiologically aims to keep serum phosphate levels within seemingly normal ranges, activates cardiomyocytes, which stepwise induces the development of LVH and subsequently of left ventricular dysfunction and congestive heart failure. While presently neither experimental nor clinical data support a direct contribution of FGF-23 in accelerated atherogenesis, phosphate loading may actively propagate vascular calcification and atherosclerosis. This may induce further myocardial injury both by increasing arterial stiffness and subsequently cardiac afterload and by hindering coronary blood supply of cardiomyocytes (compare Figure 4).

Fig. 4.

Simplified hypothetical patholophysiological concept which integrates the contribution of systemic phosphate burden and hyperphosphatoninism in the development of cardiovascular disease. Details are discussed in the text. *The selective contribution of serum phosphate to atherosclerotic plaque development is yet under discussion.

Admittedly, substantially more evidence from future clinical and experimental studies is needed to further support this hypothetical concept. Nevertheless, the persisting high cardiovascular death toll in CKD patients and the poor evidence for any beneficial intervention mandate us to conceive novel treatment strategies in CKD and to test their efficacy in clinical trials.

Three distinct therapeutic strategies for hyperphosphatoninism have been suggested: oral phosphate binders, FGF-23-blocking agents and FGF-R antagonists. Only oral phosphate binders are presently available. Their capacity to affect serum FGF-23 levels has been clearly demonstrated [86–88], and a blueprint for a large-scale, endpoint-driven clinical trial has been suggested [45]. Moreover, while oral phosphate binders are less specific agents than FGF-23-blocking agents or FGF-R antagonists, they bear the advantage of tackling two components of CKD-MBD by lowering both systemic phosphate burden and serum FGF-23. In theory, such a dual treatment strategy should allow broader vasculo- and cardioprotection.

Although FGF-23-blocking agents and FGF-R antagonists may be welcomed as modern and disease-specific drugs in hyperphosphatoninism, they bear the risk of elevating serum phosphate levels and systemic phosphate burden by inhibiting renal phosphate excretion. Of note, a better understanding of organ-specific FGF-R expression may allow to develop selective blockers of cardiac FGF-R, thus inhibiting hyperphosphatoninism-induced LVH, while leaving renal phosphate excretion unaffected.

In summary, hyperphosphatoninism has been acknowledged as a central component of CKD-MBD. A significant contribution of hyperphosphatoninism to cardiovascular disease has been suggested in large epidemiological studies and underscored by recent experimental data. Therapeutic interventions in hyperphosphatoninism should be investigated in adequately designed randomized trials.

Conflict of interest statement. S.S. has nothing to disclose. G.H.H. has been speaker for Shire. D.F. has been speaker and/or part of the advisory board for Shire, Amgen, Genzyme and FMC.

References

1
de Jager
DJ
Grootendorst
DC
Jager
KJ
, et al. 
Cardiovascular and noncardiovascular mortality among patients starting dialysis
J Am Med Assoc
2009
, vol. 
302
 (pg. 
1782
-
1789
)
2
Matsushita
K
van der Velde
M
Astor
BC
, et al. 
Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis
Lancet
2010
, vol. 
375
 (pg. 
2073
-
2081
)
3
Herzog
CA
Asinger
RW
Berger
AK
, et al. 
Cardiovascular disease in chronic kidney disease. A clinical update from Kidney Disease: Improving Global Outcomes (KDIGO)
Kidney Int
2011
, vol. 
80
 (pg. 
572
-
586
)
4
Weiner
DE
Tighiouart
H
Elsayed
EF
, et al. 
The Framingham predictive instrument in chronic kidney disease
J Am Coll Cardiol
2007
, vol. 
50
 (pg. 
217
-
224
)
5
Zannad
F
Kessler
M
Lehert
P
, et al. 
Prevention of cardiovascular events in end-stage renal disease: results of a randomized trial of fosinopril and implications for future studies
Kidney Int
2006
, vol. 
70
 (pg. 
1318
-
1324
)
6
Fellstrom
BC
Jardine
AG
Schmieder
RE
, et al. 
Rosuvastatin and cardiovascular events in patients undergoing hemodialysis
N Engl J Med
2009
, vol. 
360
 (pg. 
1395
-
1407
)
7
Wanner
C
Krane
V
Marz
W
, et al. 
Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis
N Engl J Med
2005
, vol. 
353
 (pg. 
238
-
248
)
8
Besarab
A
Bolton
WK
Browne
JK
, et al. 
The effects of normal as compared with low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin
N Engl J Med
1998
, vol. 
339
 (pg. 
584
-
590
)
9
Heinz
J
Kropf
S
Domrose
U
, et al. 
B vitamins and the risk of total mortality and cardiovascular disease in end-stage renal disease: results of a randomized controlled trial
Circulation
2010
, vol. 
121
 (pg. 
1432
-
1438
)
10
Jamison
RL
Hartigan
P
Kaufman
JS
, et al. 
Effect of homocysteine lowering on mortality and vascular disease in advanced chronic kidney disease and end-stage renal disease: a randomized controlled trial
J Am Med Assoc
2007
, vol. 
298
 (pg. 
1163
-
1170
)
11
Wrone
EM
Hornberger
JM
Zehnder
JL
, et al. 
Randomized trial of folic acid for prevention of cardiovascular events in end-stage renal disease
J Am Soc Nephrol
2004
, vol. 
15
 (pg. 
420
-
426
)
12
Cooper
BA
Branley
P
Bulfone
L
, et al. 
A randomized, controlled trial of early versus late initiation of dialysis
N Engl J Med
2010
, vol. 
363
 (pg. 
609
-
619
)
13
Eknoyan
G
Beck
GJ
Cheung
AK
, et al. 
Effect of dialysis dose and membrane flux in maintenance hemodialysis
N Engl J Med
2002
, vol. 
347
 (pg. 
2010
-
2019
)
14
Ravani
P
Malberti
F
Tripepi
G
, et al. 
Vitamin D levels and patient outcome in chronic kidney disease
Kidney Int
2009
, vol. 
75
 (pg. 
88
-
95
)
15
Tentori
F
Blayney
MJ
Albert
JM
, et al. 
Mortality risk for dialysis patients with different levels of serum calcium, phosphorus, and PTH: the Dialysis Outcomes and Practice Patterns Study (DOPPS)
Am J Kidney Dis
2008
, vol. 
52
 (pg. 
519
-
530
)
16
Wald
R
Sarnak
MJ
Tighiouart
H
, et al. 
Disordered mineral metabolism in hemodialysis patients: an analysis of cumulative effects in the Hemodialysis (HEMO) Study
Am J Kidney Dis
2008
, vol. 
52
 (pg. 
531
-
540
)
17
Wolf
M
Shah
A
Gutierrez
O
, et al. 
Vitamin D levels and early mortality among incident hemodialysis patients
Kidney Int
2007
, vol. 
72
 (pg. 
1004
-
1013
)
18
Isakova
T
Gutierrez
OM
Chang
Y
, et al. 
Phosphorus binders and survival on hemodialysis
J Am Soc Nephrol
2009
, vol. 
20
 (pg. 
388
-
396
)
19
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
, vol. 
68
 (pg. 
1815
-
1824
)
20
de Zeeuw
D
Agarwal
R
Amdahl
M
, et al. 
Selective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): a randomised controlled trial
Lancet
2010
, vol. 
376
 (pg. 
1543
-
1551
)
21
Raggi
P
Chertow
GM
Torres
PU
, et al. 
The ADVANCE study: a randomized study to evaluate the effects of cinacalcet plus low-dose vitamin D on vascular calcification in patients on hemodialysis
Nephrol Dial Transplant
2011
, vol. 
26
 (pg. 
1327
-
1339
)
22
Fukumoto
S
Physiological regulation and disorders of phosphate metabolism–pivotal role of fibroblast growth factor 23
Intern Med
2008
, vol. 
47
 (pg. 
337
-
343
)
23
Jagtap
VS
Sarathi
V
Lila
AR
, et al. 
Hypophosphatemic rickets
Indian J Endocrinol Metab
2012
, vol. 
16
 (pg. 
177
-
182
)
24
Quarles
LD
Role of FGF23 in vitamin D and phosphate metabolism: implications in chronic kidney disease
Exp Cell Res
2012
, vol. 
318
 (pg. 
1040
-
1048
)
25
Wolf
M
Fibroblast growth factor 23 and the future of phosphorus management
Curr Opin Nephrol Hypertens
2009
, vol. 
18
 (pg. 
463
-
468
)
26
Riminucci
M
Collins
MT
Fedarko
NS
, et al. 
FGF-23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting
J Clin Invest
2003
, vol. 
112
 (pg. 
683
-
692
)
27
Goetz
R
Beenken
A
Ibrahimi
OA
, et al. 
Molecular insights into the Klotho-dependent, endocrine mode of action of fibroblast growth factor 19 subfamily members
Mol Cell Biol
2007
, vol. 
27
 (pg. 
3417
-
3428
)
28
Yamashita
T
Structural and biochemical properties of fibroblast growth factor 23
Ther Apher Dial
2005
, vol. 
9
 (pg. 
313
-
318
)
29
Tohyama
O
Imura
A
Iwano
A
, et al. 
Klotho is a novel beta-glucuronidase capable of hydrolyzing steroid beta-glucuronides
J Biol Chem
2004
, vol. 
279
 (pg. 
9777
-
9784
)
30
Baum
M
Schiavi
S
Dwarakanath
V
, et al. 
Effect of fibroblast growth factor-23 on phosphate transport in proximal tubules
Kidney Int
2005
, vol. 
68
 (pg. 
1148
-
1153
)
31
Miyamoto
K
Ito
M
Kuwahata
M
, et al. 
Inhibition of intestinal sodium-dependent inorganic phosphate transport by fibroblast growth factor 23
Ther Apher Dial
2005
, vol. 
9
 (pg. 
331
-
335
)
32
Shimada
T
Hasegawa
H
Yamazaki
Y
, et al. 
FGF-23 is a potent regulator of vitamin D metabolism and phosphate homeostasis
J Bone Miner Res
2004
, vol. 
19
 (pg. 
429
-
435
)
33
Liu
S
Tang
W
Zhou
J
, et al. 
Fibroblast growth factor 23 is a counter-regulatory phosphaturic hormone for vitamin D
J Am Soc Nephrol
2006
, vol. 
17
 (pg. 
1305
-
1315
)
34
Krajisnik
T
Bjorklund
P
Marsell
R
, et al. 
Fibroblast growth factor-23 regulates parathyroid hormone and 1alpha-hydroxylase expression in cultured bovine parathyroid cells
J Endocrinol
2007
, vol. 
195
 (pg. 
125
-
131
)
35
Ben-Dov
IZ
Galitzer
H
Lavi-Moshayoff
V
, et al. 
The parathyroid is a target organ for FGF23 in rats
J Clin Invest
2007
, vol. 
117
 (pg. 
4003
-
4008
)
36
Hofman-Bang
J
Martuseviciene
G
Santini
MA
, et al. 
Increased parathyroid expression of Klotho in uremic rats
Kidney Int
2010
, vol. 
78
 (pg. 
1119
-
1127
)
37
Lopez
I
Rodriguez-Ortiz
ME
Almaden
Y
, et al. 
Direct and indirect effects of parathyroid hormone on circulating levels of fibroblast growth factor 23 in vivo
Kidney Int
2011
, vol. 
80
 (pg. 
475
-
482
)
38
Saji
F
Shigematsu
T
Sakaguchi
T
, et al. 
Fibroblast growth factor 23 production in bone is directly regulated by 1{alpha},25-dihydroxyvitamin D, but not PTH
Am J Physiol Renal Physiol
2010
, vol. 
299
 (pg. 
F1212
-
F1217
)
39
Samadfam
R
Richard
C
Nguyen-Yamamoto
L
, et al. 
Bone formation regulates circulating concentrations of fibroblast growth factor 23
Endocrinology
2009
, vol. 
150
 (pg. 
4835
-
4845
)
40
Burnett-Bowie
SM
Henao
MP
Dere
ME
, et al. 
Effects of hPTH(1–34) infusion on circulating serum phosphate, 1,25-dihydroxyvitamin D, and FGF23 levels in healthy men
J Bone Miner Res
2009
, vol. 
24
 (pg. 
1681
-
1685
)
41
Gutierrez
OM
Smith
KT
Barchi-Chung
A
, et al. 
(1–34) Parathyroid hormone infusion acutely lowers fibroblast growth factor 23 concentrations in adult volunteers
Clin J Am Soc Nephrol
2012
, vol. 
7
 (pg. 
139
-
145
)
42
Wesseling-Perry
K
Harkins
GC
Wang
HJ
, et al. 
The calcemic response to continuous parathyroid hormone (PTH)(1–34) infusion in end-stage kidney disease varies according to bone turnover: a potential role for PTH(7–84)
J Clin Endocrinol Metab
2010
, vol. 
95
 (pg. 
2772
-
2780
)
43
Larsson
T
Nisbeth
U
Ljunggren
O
, et al. 
Circulating concentration of FGF-23 increases as renal function declines in patients with chronic kidney disease, but does not change in response to variation in phosphate intake in healthy volunteers
Kidney Int
2003
, vol. 
64
 (pg. 
2272
-
2279
)
44
Gutierrez
O
Isakova
T
Rhee
E
, et al. 
Fibroblast growth factor-23 mitigates hyperphosphatemia but accentuates calcitriol deficiency in chronic kidney disease
J Am Soc Nephrol
2005
, vol. 
16
 (pg. 
2205
-
2215
)
45
Isakova
T
Gutierrez
OM
Wolf
M
A blueprint for randomized trials targeting phosphorus metabolism in chronic kidney disease
Kidney Int
2009
, vol. 
76
 (pg. 
705
-
716
)
46
Evenepoel
P
Meijers
B
Viaene
L
, et al. 
Fibroblast growth factor-23 in early chronic kidney disease: additional support in favor of a phosphate-centric paradigm for the pathogenesis of secondary hyperparathyroidism
Clin J Am Soc Nephrol
2010
, vol. 
5
 (pg. 
1268
-
1276
)
47
Isakova
T
Wahl
P
Vargas
GS
, et al. 
Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease
Kidney Int
2011
, vol. 
79
 (pg. 
1370
-
1378
)
48
Hasegawa
H
Nagano
N
Urakawa
I
, et al. 
Direct evidence for a causative role of FGF23 in the abnormal renal phosphate handling and vitamin D metabolism in rats with early-stage chronic kidney disease
Kidney Int
2010
, vol. 
78
 (pg. 
975
-
980
)
49
John
GB
Cheng
CY
Kuro-o
M
Role of Klotho in aging, phosphate metabolism, and CKD
Am J Kidney Dis
2011
, vol. 
58
 (pg. 
127
-
134
)
50
Kuro-o
M
Klotho in chronic kidney disease—what's new?
Nephrol Dial Transplant
2009
, vol. 
24
 (pg. 
1705
-
1708
)
51
Koh
N
Fujimori
T
Nishiguchi
S
, et al. 
Severely reduced production of Klotho in human chronic renal failure kidney
Biochem Biophys Res Commun
2001
, vol. 
280
 (pg. 
1015
-
1020
)
52
Marsell
R
Krajisnik
T
Göransson
H
, et al. 
Gene expression analysis of kidneys from transgenic mice expressing fibroblast growth factor-23
Nephrol Dial Transplant
2008
, vol. 
23
 (pg. 
827
-
833
)
53
Fliser
D
Kollerits
B
Neyer
U
, et al. 
Fibroblast growth factor 23 (FGF23) predicts progression of chronic kidney disease: the Mild to Moderate Kidney Disease (MMKD) Study
J Am Soc Nephrol
2007
, vol. 
18
 (pg. 
2600
-
2608
)
54
Titan
SM
Zatz
R
Graciolli
FG
, et al. 
FGF-23 as a predictor of renal outcome in diabetic nephropathy
Clin J Am Soc Nephrol
2011
, vol. 
6
 (pg. 
241
-
247
)
55
Isakova
T
Xie
H
Yang
W
, et al. 
Fibroblast growth factor 23 and risks of mortality and end-stage renal disease in patients with chronic kidney disease
J Am Med Assoc
2011
, vol. 
305
 (pg. 
2432
-
2439
)
56
Kendrick
J
Cheung
AK
Kaufman
JS
, et al. 
FGF-23 associates with death, cardiovascular events, and initiation of chronic dialysis
J Am Soc Nephrol
2011
, vol. 
22
 (pg. 
1913
-
1922
)
57
Gutierrez
OM
Mannstadt
M
Isakova
T
, et al. 
Fibroblast growth factor 23 and mortality among patients undergoing hemodialysis
N Engl J Med
2008
, vol. 
359
 (pg. 
584
-
592
)
58
Jean
G
Terrat
JC
Vanel
T
, et al. 
High levels of serum fibroblast growth factor (FGF)-23 are associated with increased mortality in long haemodialysis patients
Nephrol Dial Transplant
2009
, vol. 
24
 (pg. 
2792
-
2796
)
59
Olauson
H
Qureshi
AR
Miyamoto
T
, et al. 
Relation between serum fibroblast growth factor-23 level and mortality in incident dialysis patients: are gender and cardiovascular disease confounding the relationship?
Nephrol Dial Transplant
2010
, vol. 
25
 (pg. 
3033
-
3038
)
60
Seiler
S
Reichart
B
Roth
D
, et al. 
FGF-23 and future cardiovascular events in patients with chronic kidney disease before initiation of dialysis treatment
Nephrol Dial Transplant
2010
, vol. 
25
 (pg. 
3983
-
3989
)
61
Nakano
C
Hamano
T
Fujii
N
, et al. 
Intact fibroblast growth factor 23 levels predict incident cardiovascular event before but not after the start of dialysis
Bone
2012
, vol. 
50
 (pg. 
1266
-
1274
)
62
Dhingra
R
Sullivan
LM
Fox
CS
, et al. 
Relations of serum phosphorus and calcium levels to the incidence of cardiovascular disease in the community
Arch Intern Med
2007
, vol. 
167
 (pg. 
879
-
885
)
63
Larsson
TE
Olauson
H
Hagstrom
E
, et al. 
Conjoint effects of serum calcium and phosphate on risk of total, cardiovascular, and noncardiovascular mortality in the community
Arterioscler Thromb Vasc Biol
2010
, vol. 
30
 (pg. 
333
-
339
)
64
Tonelli
M
Curhan
G
Pfeffer
M
, et al. 
Relation between alkaline phosphatase, serum phosphate, and all-cause or cardiovascular mortality
Circulation
2009
, vol. 
120
 (pg. 
1784
-
1792
)
65
Tonelli
M
Sacks
F
Pfeffer
M
, et al. 
Relation between serum phosphate level and cardiovascular event rate in people with coronary disease
Circulation
2005
, vol. 
112
 (pg. 
2627
-
2633
)
66
Dobnig
H
Pilz
S
Scharnagl
H
, et al. 
Independent association of low serum 25-hydroxyvitamin d and 1,25-dihydroxyvitamin d levels with all-cause and cardiovascular mortality
Arch Intern Med
2008
, vol. 
168
 (pg. 
1340
-
1349
)
67
Parker
BD
Schurgers
LJ
Brandenburg
VM
, et al. 
The associations of fibroblast growth factor 23 and uncarboxylated matrix Gla protein with mortality in coronary artery disease: the Heart and Soul Study
Ann Intern Med
2010
, vol. 
152
 (pg. 
640
-
648
)
68
Taylor
EN
Rimm
EB
Stampfer
MJ
, et al. 
Plasma fibroblast growth factor 23, parathyroid hormone, phosphorus, and risk of coronary heart disease
Am Heart J
2011
, vol. 
161
 (pg. 
956
-
962
)
69
Semba
RD
Fink
JC
Sun
K
, et al. 
Serum fibroblast growth factor-23 and risk of incident chronic kidney disease in older community-dwelling women
Clin J Am Soc Nephrol
2012
, vol. 
7
 (pg. 
85
-
91
)
70
Udell
JA
O'Donnell
T
Morrow
D
, et al. 
Association of fibroblast growth factor (FGF)-23 levels with risk of cardiovascular events in patients with stable coronary artery disease
J Am Coll Cardiol
2012
, vol. 
59
 pg. 
E1480
 
71
Seiler
S
Cremers
B
Rebling
NM
, et al. 
The phosphatonin fibroblast growth factor 23 links calcium-phosphate metabolism with left-ventricular dysfunction and atrial fibrillation
Eur Heart J
2011
, vol. 
32
 (pg. 
2688
-
2696
)
72
Ellam
T
Wilkie
M
Chamberlain
J
, et al. 
Dietary phosphate modulates atherogenesis and insulin resistance in apolipoprotein E knockout mice–brief report
Arterioscler Thromb Vasc Biol
2011
, vol. 
31
 (pg. 
1988
-
1990
)
73
Jono
S
McKee
MD
Murry
CE
, et al. 
Phosphate regulation of vascular smooth muscle cell calcification
Circ Res
2000
, vol. 
87
 (pg. 
E10
-
E17
)
74
Neves
KR
Graciolli
FG
dos Reis
LM
, et al. 
Adverse effects of hyperphosphatemia on myocardial hypertrophy, renal function, and bone in rats with renal failure
Kidney Int
2004
, vol. 
66
 (pg. 
2237
-
2244
)
75
Portale
AA
Halloran
BP
Morris
RC
Jr
Dietary intake of phosphorus modulates the circadian rhythm in serum concentration of phosphorus. Implications ofr the renal production of 1,25-dihydroxyvitamin D
J Clin Invest
1987
, vol. 
80
 (pg. 
1147
-
1154
)
76
American Diabetes Association. Standards of medical care in diabetes—2009
Diabetes Care
2009
, vol. 
32
 
Suppl 1
(pg. 
S13
-
S61
)
77
Faul
C
Fibroblast growth factor 23 and the heart
Curr Opin Nephrol Hypertens
2012
, vol. 
21
 (pg. 
369
-
375
)
78
Gutierrez
OM
Januzzi
JL
Isakova
T
, et al. 
Fibroblast growth factor 23 and left ventricular hypertrophy in chronic kidney disease
Circulation
2009
, vol. 
119
 (pg. 
2545
-
2552
)
79
Mirza
MA
Larsson
A
Melhus
H
, et al. 
Serum intact FGF23 associate with left ventricular mass, hypertrophy and geometry in an elderly population
Atherosclerosis
2009
, vol. 
207
 (pg. 
546
-
551
)
80
Faul
C
Amaral
AP
Oskouei
B
, et al. 
FGF23 induces left ventricular hypertrophy
J Clin Invest
2011
, vol. 
121
 (pg. 
4393
-
4408
)
81
Frohlich
ED
Apstein
C
Chobanian
AV
, et al. 
The heart in hypertension
N Engl J Med
1992
, vol. 
327
 (pg. 
998
-
1008
)
82
Levy
D
Larson
MG
Vasan
RS
, et al. 
The progression from hypertension to congestive heart failure
J Am Med Assoc
1996
, vol. 
275
 (pg. 
1557
-
1562
)
83
Ellam
TJ
Chico
TJ
Phosphate: the new cholesterol? The role of the phosphate axis in non-uremic vascular disease
Atherosclerosis
2012
, vol. 
220
 (pg. 
310
-
318
)
84
Roos
M
Lutz
J
Salmhofer
H
, et al. 
Relation between plasma fibroblast growth factor-23, serum fetuin-A levels and coronary artery calcification evaluated by multislice computed tomography in patients with normal kidney function
Clin Endocrinol (Oxf)
2008
, vol. 
68
 (pg. 
660
-
665
)
85
Mirza
MA
Hansen
T
Johansson
L
, et al. 
Relationship between circulating FGF23 and total body atherosclerosis in the community
Nephrol Dial Transplant
2009
, vol. 
24
 (pg. 
3125
-
3131
)
86
Koiwa
F
Kazama
JJ
Tokumoto
A
, et al. 
Sevelamer hydrochloride and calcium bicarbonate reduce serum fibroblast growth factor 23 levels in dialysis patients
Ther Apher Dial
2005
, vol. 
9
 (pg. 
336
-
339
)
87
Nagano
N
Miyata
S
Abe
M
, et al. 
Effect of manipulating serum phosphorus with phosphate binder on circulating PTH and FGF23 in renal failure rats
Kidney Int
2006
, vol. 
69
 (pg. 
531
-
537
)
88
Pande
S
Ritter
CS
Rothstein
M
, et al. 
FGF-23 and sFRP-4 in chronic kidney disease and post-renal transplantation
Nephron Physiol
2006
, vol. 
104
 (pg. 
p23
-
p32
)

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.