Skip to main content
Clinical Journal of the American Society of Nephrology : CJASN logoLink to Clinical Journal of the American Society of Nephrology : CJASN
. 2014 Feb 27;9(7):1283–1303. doi: 10.2215/CJN.10941013

The Use of Fibroblast Growth Factor 23 Testing in Patients with Kidney Disease

Edward R Smith 1,
PMCID: PMC4078965  PMID: 24578336

Abstract

The emergence of fibroblast growth factor 23 as a potentially modifiable risk factor in CKD has led to growing interest in its measurement as a tool to assess patient risk and target therapy. This review discusses the analytical and clinical challenges faced in translating fibroblast growth factor 23 testing into routine practice. As for other bone mineral markers, agreement between commercial fibroblast growth factor 23 assays is poor, mainly because of differences in calibration, but also, these differences reflect the variable detection of hormone fragments. Direct comparison of readout from different assays is consequently limited and likely hampers setting uniform fibroblast growth factor 23–directed targets. Efforts are needed to standardize assay output to enhance clinical use. Fibroblast growth factor 23 is robustly associated with cardiovascular and renal outcomes in patients with CKD and adds value to risk assessments based on conventional risk factors. Compared with most other mineral markers, fibroblast growth factor 23 shows better intraindividual temporal stability, with minimal diurnal and week-to-week variability, but substantial interindividual variation, maximizing discriminative power for risk stratification. Conventional therapeutic interventions for the CKD–mineral bone disorder, such as dietary phosphate restriction and use of oral phosphate binders or calcimimetics, are associated with variable efficacy at modulating circulating fibroblast growth factor 23 concentrations, like they are for other mineral metabolites. Dual therapy with dietary phosphate restriction and noncalcium-based binder use achieves the most consistent fibroblast growth factor 23–lowering effect and seems best monitored using an intact assay. Additional studies are needed to evaluate whether strategies aimed at reducing levels or antagonizing its action have beneficial effects on clinical outcomes in CKD patients. Moreover, a better understanding of the mechanisms driving fibroblast growth factor 23 elevations in CKD is needed to inform the use of therapeutic interventions targeting fibroblast growth factor 23 excess. This evidence must be forthcoming to support the use of fibroblast growth factor 23 measurement and fibroblast growth factor 23–directed therapy in the clinic.

Keywords: FGF23, CKD-MBD, ELISA, cardiovascular disease, risk factors

Introduction

Over the last decade, fibroblast growth factor 23 (FGF23) has emerged as one of the main bone-derived physiologic endocrine regulators of mineral metabolism in vertebrates, chiefly functioning as a counter-regulatory hormone for 1,25 dihydroxyvitamin D [1,25(OH)2D] (1,2). The recognition that circulating FGF23 concentrations rise early in the progression of CKD and are not only strongly predictive of cardiovascular and renal end points (36) but, in particular, seem mechanistically linked to pathology (7) has driven optimism that earlier institution of therapies targeting FGF23 excess might improve patient outcomes. However, before FGF23 measurement can be incorporated into the standard laboratory testing repertoire and considered a bone fide clinical tool, it must be shown to have met key analytical and clinical benchmarks. The focus of this review is to appraise three such benchmarks: (1) ready availability of accurate and reproducible FGF23 assays suitable for testing in the modern clinical laboratory, (2) evidence that FGF23 measurement adds to or improves on existing testing strategies, and (3) evidence that the risk associated with FGF23 is modifiable or may help guide patient care.

Analytical Considerations of FGF23 Measurement

Intact FGF23 (approximately 32 kDa) contains a proteolytic cleavage site (R176XXR179) recognized by a family of serine endopeptidase proprotein convertases (8). Cleavage at this locus generates N- and C-terminal fragments (approximately 18 and 14 kDa, respectively) that, with intact hormone, enter the circulation (9). Two formats of immunometric FGF23 assay are available from several commercial manufacturers. Intact FGF23 (iFGF23) ELISAs, developed by Immutopics, Inc., Kainos Laboratories, and EMD Millipore, require simultaneous recognition of epitopes within the N- and C-terminal domains flanking the proteolytic cleavage site and detect only full-length hormone (10). Although different iFGF23 assays yield closely correlated results, they show poor absolute numerical agreement because of calibration differences (11,12). iFGF23 assays are also variably affected by instability issues caused by ex vivo proteolytic degradation of intact hormone, necessitating the collection of protease-protected plasma samples in circumstances where sample processing is delayed (>2 hours) (13).

The C-terminal FGF23 (cFGF23) ELISA from Immutopics, Inc., uses antibodies that bind two distinct epitopes within the C-terminal portion of the protein, thus detecting both intact and C-terminal fragments (14). The most recent generation of the cFGF23 assay seems least affected by preanalytical stability issues (11). It is important to recognize, however, that cFGF23 measurements do not necessarily reflect changes in FGF23 bioactivity, particularly in situations where FGF23 processing is altered. Patients with iron-deficiency anemia (15), fibrous dysplasia (16), or familial tumoral calcinosis (17) can display elevated cFGF23 concentrations, whereas intact hormone is normal or even low. The biologic activity of cFGF23 fragments remains a controversial topic. Early work by Berndt et al. (18) suggested that proteolytically generated peptides containing residues 180–205 retained the phosphaturic activity of intact hormone in opossum kidney cells but that further truncation of this domain abolished such activity. Goetz et al. (19) later localized the minimal receptor binding epitope to this region but contrastingly, provided in vitro and in vivo evidence that the isolated cFGF23 domain dose-dependently antagonized the activity of the intact hormone by competing for occupancy of the receptor complex. In general, therefore, iFGF23 measurements represent a more homogenous signal and are likely to provide the most reliable readout of biologic effect.

iFGF23 and cFGF23 assays often show poor analytical agreement, particularly at near-physiologic concentrations (20). This discordance relates not only to calibration differences but also, the detection of C-terminal fragments normally present in plasma using the cFGF23 assay (20). Formally defined reference intervals for iFGF23 (12–49 pg/ml) and cFGF23 (22–91 RU/ml) assays have been published for adult and pediatric populations (20,21) but not for all commercial kits.

In health, iFGF23 shows significant diurnal variability (approximately 30%), with concentrations generally reaching their peak in the early morning (20,22). cFGF23 levels show minimal circadian cycling (20,23,24) and less week-to-week intraindividual variability than other conventional mineral parameters (20,25,26). Conversely, although interindividual variability for iFGF23 and parathyroid hormone (PTH) concentrations seems similar, interindividual variation for cFGF23 is markedly higher (20). This yields a high index of individuality for cFGF23 and, although helpful for risk stratification, imposes limits on clinical interpretation with conventional population-based reference intervals (27). The relatively high intraindividual variation of PTH and phosphate means that, although serial cFGF23 concentrations need to change by only 25% in an individual to be considered significant, PTH and phosphate levels need to change by more than 32% and 56%, respectively (20). In contrast, in patients undergoing hemodialysis, the intraindividual variation of plasma cFGF23 concentrations seems higher than for other bone mineral markers according to some (28,29) but not all reports (30).

Recently, Shimizu et al. (31) developed a fully automated chemiluminescent immunoassay using the monoclonal antibodies from the Kainos iFGF23 ELISA. Compared with commercial ELISAs, this assay boasts better analytical sensitivity, greater analytical range, shorter assay time, and it is relatively sample-sparing; thus, it seems better suited to a high-throughput clinical laboratory. EMD Millipore has also released a Luminex-based multiplex immunoassay bone panel including FGF23, offering the opportunity for simultaneous measurement of FGF23 and other bone and inflammatory biomarkers (e.g., PTH, sclerostin, and TNF-α) in a single sample aliquot. However, despite having the broadest working range of available assays and thus, diminishing the need for repeat testing at dilution in patients with high concentrations, the analytical limit of detection of this assay is relatively high (within the physiologic range), and the manufacturer’s performance data indicate quite high interassay imprecision. The application of these newer analytical platforms to clinical cohorts is awaited, and independent validation is required.

Table 1 compares key performance characteristics of available FGF23 assays. Currently, commercial FGF23 assays have not been approved or validated for in vitro diagnostic use. Overall, marked calibration differences constrain direct interassay comparisons. It should be stressed, however, that problems with assay standardization are not unique to FGF23, and similar long-standing issues are well described for many other bone mineral markers, most notably PTH and 25 hydroxyvitamin D [25(OH)D], that have been measured routinely for decades (32,33). Nonetheless, such experience with other mineral metabolism analytes should prompt attempts to harmonize FGF23 assay readout.

Table 1.

Performance characteristics of assays for FGF23 measurement

Characteristic Intact FGF23 C-Terminal FGF23 (Immutopics, Inc.a)
Kainos Laboratories Immutopics, Inc.a EMD Millipore Shimizu et al. (31) EMD Millipore
Type/detection ELISA/colorimetric ELISA/colorimetric ELISA/colorimetric CLIA/chemiluminescent FIA Luminex/fluorescent ELISA/colorimetric
Epitope binding region Unknown Residues 51–69 Unknown Unknown Unknown Residues 206–222
Residues 206–222 Residues 225–244
Preferred sample type Serum EDTA plasma EDTA plasma or serum Serum EDTA plasma or serum EDTA plasma
Preanalytical stability issues No Yes (13) No Not tested Not tested No
Sample volumeb (μl) 100 100 100 20 50 100
Functional sensitivity 2 pg/ml 1.5 pg/ml 18 pg/ml 1 pg/ml 37 pg/ml 1.5 RU/ml
Analytical range 2–800 pg/ml 2–2200 pg/ml 18–2400 pg/ml 1–15,000 pg/ml 37–150,000 pg/ml 2–1400 RU/ml
Interassay imprecision (%) <5 <5 <12 <5 <15 <4
Assay time 3–4 h 3–4 h 3–4 h 20 min 2–16 h 3–4 h
Multiplex capability No No No No Yes No
Independent analytical validation Yes (11,140) Yes for first generation kit (11,140) Yes (11) No No Yes (11,140)
Reference intervalc No Yes, for first generation kit (20) No No No Yes (20,21)
Clinical outcome studiesd Multiple (36,42,46,50,74,75) Several (43,141) None None None Multiple (36,55,59,60,142)

FGF23, fibroblast growth factor 23; CLIA, chemiluminescence immunoassay; FIA, fluorescence immunoassay.

a

Second generation kits.

b

For duplicate determinations.

c

Defined according to Clinical and Laboratory Standards Institute guidelines (143).

d

Death or progression to ESRD requiring RRT.

Does FGF23 Measurement Add Value for Patients with CKD?

CKD as a State of FGF23 Excess

According to analysis of 3879 patients with stages 2–4 CKD in the Chronic Renal Insufficiency Cohort (CRIC) study (3), plasma cFGF23 concentrations increase early in disease (eGFR=58 ml/min per 1.73 m2), before elevations in PTH (eGFR=47 ml/min per 1.73 m2), and consistent with earlier studies (34), well before the onset of frank hyperphosphatemia (<30 ml/min per 1.73 m2). In CRIC, few patients (<6%) showed an isolated increase in PTH (>65 pg/ml but normal FGF23), emphasizing the primacy of elevations in FGF23 in this patient group (3). Consistent findings have been reported in pediatric CKD cohorts using the same assay (35) and other adult nondialysis-dependent CKD cohorts using the Kainos iFGF23 assay (36). This temporal sequence is also borne out in longitudinal studies in murine models of progressive CKD (37). FGF23 concentrations increase inexorably as renal function falls, where intact full-length hormone seems to be the predominant circulating species (20,38). In dialysis patients without residual renal function, concentrations can be 1000-fold above normal (39,40), and after kidney transplantation, FGF23 levels decline rapidly in patients with good allograft function (41).

FGF23 and Adverse Outcomes in CKD

Multiple prospective studies have shown a consistent link between higher FGF23 concentrations and increased risk of both death and cardiovascular events across the entire spectrum of renal disease (4,5,4245). Most studies show a near-linear increase in risk with increasing FGF23 concentrations, with available (but limited) data suggesting comparable results between cFGF23 and iFGF23 measurements (43). These relationships seem minimally confounded by renal risk factors, conventional cardiovascular risk factors, therapeutic interventions, or, perhaps most significantly, other mineral parameters as well. Even adjustment for measured GFR does not seem to materially attenuate the association (4,46). As summarized in Table 2, after full adjustment, effects remain sizeable and stable across a number of diverse and well powered observational CKD cohort studies. Conversely, substitution of FGF23 for other mineral exposures, including PTH, 25(OH)D, 1,25(OH)2D, or fractional excretion of phosphate (FEP), in the same multivariate models has failed to consistently show the same monotonic relationship with outcome (Table 3) (4,5,42), suggesting an effect specific to FGF23 rather than a more general one reflecting mineral disturbances. Indeed, in CRIC and the Homocysteine in Kidney and End Stage Renal Disease studies, the mortality risk associated with FGF23 is reportedly even greater than for established CKD-specific factors, like eGFR and proteinuria (4,5). Hence, the magnitude of the risk of hard end points with very high FGF23 concentrations suggests that FGF23 may well present an important clinical target in such patients in the future. Incorporation of FGF23 measurements into cardiovascular risk models based on conventional risk factors (e.g., Framingham risk factors) seems to yield a small but significant improvement in risk reclassification in both nondialysis-dependent CKD (7%) and post-transplant (7%–11%) cohorts (42,45).

Table 2.

Association of FGF23 concentration with all-cause mortality and cardiovascular events in large prospective nondialysis-dependent CKD cohort studies

Study Demographics: n; Age (yr); Sex (% Men); Race Renal Function (CKD; eGFR) FGF23 Assay/Median Level (IQR) Outcome Events (n, %)/Median Follow-Up (yr) Adjusted Hazard Ratio (95% CI)a
Continuous Highest Versus Lowest Group
Isakova et al. (CRIC) (4) 3879; 58±11; 56; 42% African American 2–4; 43±14 cFGF23/146 (96–239) All-cause mortality 266, 7/3.5 1.5 (1.3 to 1.7) per 1 SD included 3.0 (1.8 to 5.1)
Scialla et al. (CRIC) (6) 3860; 58±11; 56; 42% African American 2–4; 44±15 cFGF23/145 (96–239) Atherosclerotic events; CHF events 287, 7; 360, 9/3.6 1.2 (1.1 to 1.4) per doubling; 1.5 (1.3 to 1.7) per doubling 1.8 (1.2 to 2.6); 3.0 (2.0 to 4.5)
Kendrick et al. (HOST) (5) 1099; 69±11; 98; 26% African American 4–5; 18±16 cFGF23/392 (216–945) All-cause mortality 453, 41/2.9 1.6 (1.3 to 2.1) per 1 SD included 2.2 (1.6 to 3.1)
Scialla et al. (AASK) (46) 809; 55±10; 60; 100% African American 2–5; 45±18 (125I-iothalamate) iFGF23/44 (31–64) Composite of all-cause mortality and initiation of RRT 351, 43 (117, 14 died)/7.9 1.3 (1.2 to 1.5) per doubling 2.3 (1.3 to 3.8)
Nakano et al. (OVIDS-CKD) (42) 738; 64 (54–72); 66; Japanese 1–5; 35±19 iFGF23/50 (32–81) CV events predialysis 215, 29/4.4 1.7 (1.1 to 2.7) per 1 SD included Not reported
Wolf et al. (44) 984; 55±10; 57; European Post-transplant;51±21 cFGF23/28 (20–43) Composite of all-cause mortality and allograft loss 182, 18; 87, 9 died/3.1 1.4 (1.3 to 1.7) per 1 SD included; 1.5 (1.3 to 1.8) per 1 SD included 2.2 (1.4 to 3.7); 2.2 (1.1 to 4.0)
Baia et al. (45) 593; 52±12; 54; European (95% white) Post-transplant;47±16 cFGF23/140 (95–219) CV mortality; all-cause mortality 66, 11; 128, 22/7.0 1.9 (1.1 to 3.2) per 1 SD included; 1.9 (1.3 to 2.7) per 1 SD included 3.0 (1.1 to 8.3); 2.2 (1.1 to 4.5)

IQR, interquartile range; 95% CI, 95% confidence interval; CRIC, Chronic Renal Insufficiency Cohort; cFGF23, C-terminal FGF23; CHF, congestive heart failure; HOST, Homocysteine in Kidney and End Stage Renal Disease; AASK, African American Study of Kidney Disease and Hypertension; iFGF23, intact FGF23; OVIDS-CKD, Osaka Vitamin D Study in Patients with CKD; CV, cardiovascular.

a

Hazard ratios for fully adjusted models (exact model covariates differ but include adjustment for potential conventional renal, CV, and mineral confounders).

Table 3.

Independent association of conventional mineral markers with all-cause mortality and CV events in large prospective nondialysis-dependent CKD cohort studies

Study End Point Phosphate PTH 25(OH)D 1,25(OH)2D FEP
CRIC (4,6) All-cause mortality NR No NA NA No
CRIC (4,6) Atherosclerotic events Yes; HR, 1.1 (95% CI, 1.0 to 1.2) per 0.5 mg/dl No NA NA No
CRIC (4,6) Heart failure events Yes; HR, 1.1 (95% CI, 1.0 to 1.2) per 0.5 mg/dl No NA NA NA
HOST (5,56) All-cause mortality No No No No NA
HOST (5,56) CV events Yes; HR, 1.2 (95% CI, 1.1 to 1.3) per mg/dl No No Not independent of FGF23 NA
AASK (46) All-cause mortality and initiation of RRT (composite) Only at high levels>3.5 mg/dl; HR, 1.3 (95% CI, 1.2 to 1.5) per 0.5 mg/dl; not significant in tertile analysis Only at high levels>39 pg/ml; HR, 1.3 (95% CI, 1.1 to 1.5) per doubling; not significant in tertile analysis No NA NA
OVIDS-CKD (42) CV events predialysis NR No No; predictive of death but not independently of hemoglobin No NA
Wolf et al. (44) All-cause mortality and allograft loss (composite) Yes; HR, 1.2 (95% CI, 1.1 to 1.4) per 1 SD; not significant in tertile analysis or patients with eGFR=30–90 ml/min per 1.73 m2 No NA NA NA
Baia et al. (45) CV mortality No No No No No

CV, cardiovascular; PTH, parathyroid hormone; 25(OH)D, 25 hydroxyvitamin D; 1,25(OH)2D, 1,25 dihydroxyvitamin D; FEP, fractional excretion of phosphate; NR, not reported; NA, not assessed; HR, hazard ratio.

With respect to renal outcomes, higher FGF23 concentrations have been associated with earlier initiation of chronic dialysis and significant decrements in renal function in patients with nondialysis-dependent CKD (4,5,36,46), again yielding improvements in event reclassification (19%) (47), and have variably predicted graft failure after kidney transplantation (44,45). Moreover, FGF23 predicts progression in disease-specific causes of renal failure, including IgA nephropathy, and those patients with heavy proteinuric diabetic kidney disease (48,49). Recently, Semba et al. (50) also reported an independent association between FGF23 concentrations and the risk of incident CKD in a cohort of elderly community-dwelling women.

In the acute setting, FGF23 concentrations are reportedly much higher in patients with AKI compared with controls (more than 5-fold), with higher levels being predictive of greater risk of death or requirement for dialysis (51). Other mineral parameters were not predictive of outcome. Furthermore, in analysis of 3241 participants from the Cardiovascular Health Study (CHS) (52), an elderly community-dwelling cohort mostly free from renal disease (mean eGFR=71 ml/min per 1.73 m2), those individuals in the highest cFGF23 quartile had an almost 2-fold higher risk of future AKI hospitalization (median 10-year follow-up) compared with those individuals in the lowest quartile after full adjustment for renal (baseline eGFR and albuminuria) and cardiovascular risk factors.

The added value of FGF23 measurements to conventional renal and cardiovascular risk factors is noteworthy, because risk prediction models derived from the general population generally perform poorly in CKD patients (53). On this basis, a strong argument for the assimilation of FGF23 measurements into standard risk prediction models could be made. Importantly, the association between FGF23 and outcomes does seem modified by a number of factors, most notably, vitamin D status and FEP (5456). Those patients with both high FGF23 and low 25(OH)D/1,25(OH)2D or high FGF23 and low FEP seem to have the greatest risks of left ventricular remodeling (54), poor renal survival, and cardiovascular events (55). Thus, these measures may provide complementary information, and the use of biomarker panels may enhance risk stratification further. However, these findings need to be replicated in other studies.

FGF23—Direct Link to Pathology?

Although studies in animal CKD models attest to a protective role in early CKD (57), possibly as a long-term but indirect and adaptive response to a high-phosphate load (58), other evidence alludes to potentially harmful effects of chronic FGF23 excess on various organ systems, most notably the heart (7).

Accumulating epidemiologic evidence now points strongly to a relationship between FGF23 and cardiac failure rather than occlusive atherosclerotic vascular disease (6). Indeed, even in cohorts without significant renal dysfunction, such as the Heart and Soul Study (59) and the CHS (60), a similar pattern is manifest, with the association between FGF23 and death mainly driven by heart failure events rather than myocardial infarction or cerebrovascular injury. Likewise, in patients undergoing coronary angiography, again with minimal renal dysfunction (19% eGFR<60 ml/min per 1.73 m2), FGF23 has primarily been associated with left ventricular dysfunction (61). Moreover, although higher FGF23 concentrations have been associated with numerous intermediate markers of vascular disease in both adult and pediatric CKD cohorts, the most consistent associations by far are with left ventricular mass index (LVMI) and left ventricular hypertrophy (LVH) (7,6165). Analysis of CRIC shows not only a strong independent association between FGF23 and LVMI and the prevalence of LVH but also, the ability of FGF23 levels to predict incident LVH as well (7). Given the high prevalence of LVH in CKD and its link to congestive heart disease, arrhythmia, and sudden cardiac death (66), all of which contribute appreciably to the high burden of cardiovascular death in CKD (67), this finding represents a particularly important potential pathway of risk in this population. Hence, of great relevance to these epidemiologic observations were the landmark findings of Faul et al. (7), showing that FGF23 can directly induce hypertrophic gene programs in the cardiomyocyte, leading to LVH at the elevated levels seen in some advanced CKD patients. Independent investigators have subsequently corroborated these finding, showing the hypertrophic effect of FGF23 on cultured cardiomyocytes, and suggested that FGF23 might also have additional effects on myocardial contractility (68). Notably, in the study by Faul et al. (7), these effects were apparently transduced by the FGF receptor independent of α-klotho, which, according to the current paradigm, is an obligate coreceptor for physiologic FGF23 signaling in the kidney tubules and parathyroid glands (69). Indeed, as much as CKD would seem to be a state of FGF23 excess, CKD could also be characterized as state of α-klotho deficiency, with parathyroid and particularly, renal expression being markedly downregulated in early disease (70,71). This finding may be significant, because α-klotho deficiency could theoretically potentiate the effects of chronic FGF23 excess in advanced CKD by favoring nonspecific, low-affinity FGF receptor activation in nonphysiologic tissues, like the heart, but also feedback to the osteocyte, driving further increments in production to maintain biologic effect.

Although the aforementioned effects of FGF23 on cardiomyocytes provide a credible explanation of how very high FGF23 concentrations might drive cardiac dysfunction in dialysis patients, it is difficult to attribute the same effect to concentrations only fractionally above normal levels (72,73). Indeed, FGF23 concentrations have been robustly associated with all-cause mortality and cardiovascular events in a number of community-dwelling elderly cohorts largely free from significant renal dysfunction (59,60,74,75). Thus, additional work is required to understand the nature of the increased risk in individuals with normal or minimally reduced renal function and determine whether FGF23 might be causally related to cardiovascular disease in this setting or whether FGF23 levels are providing a surrogate marker of other processes directly related to pathology.

Although FGF23 levels have also been fairly consistently associated with poor renal end points, there is no mechanistic evidence linking higher FGF23 with renal disease progression. Nonetheless, the fact that TGF-β and TNF-α pathways are activated in murine CKD models after the administration of exogenous FGF23 (76) invokes FGF23 as a possible protagonist in renal fibrosis and inflammation. Moreover, in human studies, higher FGF23 concentrations have been independently associated with systemic proinflammatory cytokine levels (IL-6 and TNF-α) (77), suggesting a role in inflammatory processes and providing another plausible but unexplored risk pathway through which FGF23 could operate.

Rise of FGF23 in CKD—Unanswered Questions

The trigger for FGF23 elevations in CKD and the etiology of the prodigious increase in levels as renal function fails are poorly understood. Although multiple factors in advanced CKD favor increased synthetic drive (hyperparathyroidism, iron deficiency, inflammation, active vitamin D therapy, acidosis, hypercalemia, and feedback from end organ resistance caused by α-klotho downregulation) (78), osteocytic FGF23 gene expression and protein levels seem only modestly increased (if at all) in early human CKD and animal models (37,79,80), and they do not seem commensurately increased with circulating levels in dialysis patients (79). Indeed, in longitudinal analysis of an early progressive CKD rodent model, the rise in circulating FGF23 concentrations preceded changes in FGF23 gene expression in bone by 4 weeks (37). The source of circulating FGF23, particularly in early CKD, would, therefore, seem uncertain. Theoretically, very rapid changes in FGF23 levels, which were recently reported in mice with folic acid-induced AKI (81), may reflect the release of preformed hormone from bone matrix rather than de novo cellular synthesis of new protein. An alternative explanation of these data is that extraskeletal production of FGF23 may also contribute to the circulating pool. Although some analyses clearly identify bone as the principal source of FGF23 in uremic and nonuremic rodent models (82,83), low-level expression in human tissue has also been variously reported in the liver, thymus, lymph nodes, spleen, small intestine, kidney, and heart (8486). Whether CKD might affect the expression of FGF23 in these tissues and whether local production, possibly as a compensatory response to tissue injury, significantly augments systemic hormone levels seem worthy of additional investigation. In this regard, it seems particularly noteworthy that renal FGF23 expression has been reported in rodent models of diabetic nephropathy and polycystic kidney disease (87,88).

Little is known about FGF23 degradation and clearance. A growing number of studies suggest that renal clearance contributes minimally to FGF23 elimination (39,89), and unlike other peptide hormones, the high levels in CKD do not seem to be caused by accumulation of inactive hormone fragments (20,38). Additional clues come from the study of murine AKI, where renal impairment was found to only have a relatively modest effect on plasma FGF23 half-life (approximately 50% reduction) (81). The fact that neither increased osteocytic synthesis nor reduced renal clearance seems to account for the high levels typically seen in CKD suggests that extraskeletal sites of FGF23 synthesis and/or extrarenal routes of elimination may be important determinants in the evolution of FGF23 levels in this setting. Although controversial, a clear understanding of mechanisms underlying the increase in FGF23 in kidney disease is vital to fully comprehend the significance of changes in measured concentrations and better inform the use of therapeutic interventions targeting FGF23 excess.

FGF23—a Modifiable Risk Factor?

The strong association between FGF23 and mortality and the evidence that FGF23 excess may directly harm the heart raise the fundamental question of whether lowering FGF23 concentrations or antagonizing its action would reduce risk and improve patient outcome. Indeed, can conventional CKD–mineral bone disorder (CKD-MBD) therapy be used to successfully target FGF23?

Dietary Phosphate Restriction and Oral Phosphate Binders

Lowering FGF23 by restricting dietary phosphate content or reducing intestinal absorption with oral phosphate binders (or a combination of the two) has been the most comprehensively evaluated conventional CKD-MBD approach to date, but it has yielded somewhat inconsistent results, particularly in randomized controlled trials of nondialysis-dependent CKD patients (Table 4). To some extent, the apparent discordance is likely to reflect a number of factors from differences in binder type or dietary control/content to the duration of follow-up, sample size, and the FGF23 assay. The source of dietary phosphate seems to have quite a profound effect on changes in FGF23, at least in the short-term, with meat-based protein diets yielding higher levels than plant-based protein diets (despite equivalent phosphate content) (90). Dietary phosphate restriction strategies alone have yielded very modest nonsignificant reductions in FGF23 (9194), although quite extreme restriction with a very low-protein diet (<400 mg phosphate/d) has been associated with a significant reduction in circulating levels (92). Overall, reductions in FGF23 have been seen most consistently in dialysis cohorts and treatment groups receiving noncalcium-based binders. Of note, the use of calcium-based phosphate binders is theoretically undesirable given the stimulating effect of calcium on FGF23 synthesis (95), which might negate any beneficial effect on phosphate balance in addition to possibly promoting extraosseous mineral accrual (96). Assessment of FGF23 modulation using the cFGF23 assay has registered mostly null results and may be relatively insensitive to changes in bioactive levels (97100). Combined strategies of dietary phosphate restriction (<700–750 mg/d) in addition to an oral binder seem to be more effective in achieving significant reductions in FGF23 than either approach in isolation (91,93,94), but the magnitude of this decrease is quite variable and not observed consistently in all patients on treatment (like for serum phosphate concentration and urinary phosphate excretion). However, the inconsistent response to such therapeutic interventions may also simply reflect the modest and variable effect that dietary phosphate has on FGF23 in CKD. Indeed, in an analysis of 880 individuals from the Heart and Soul Study (mean eGFR=71±22 ml/min per 1.73 m2), after adjustment for demographic factors and eGFR, Palomino et al. (101) failed to detect any significant association between 24-hour urinary phosphate excretion (a measure of dietary phosphate intake) and cFGF23 concentrations. Conversely, Gutierrez et al. (102) did report a significant association between cFGF23 and dietary phosphate intake (assessed by food logs) in 1261 individuals enrolled in the Health Professionals follow-up study. However, the magnitude of this effect was still modest: every 500-mg/d increase in dietary phosphate intake was associated with only a 3.4-RU/ml increase in cFGF23 (102). Even data from animal CKD models are unpersuasive, particularly in early progressive models of disease. Some studies either show a lack of response to phosphate restriction altogether (80) or relate improvement in cardiovascular function to changes in phosphate rather than FGF23 (103). While extreme, phosphate restriction (zero phosphate diet), however, has been associated with a paradoxical increase in plasma FGF23 (104).

Table 4.

Responsiveness of FGF23 concentration to oral phosphate binder use in randomized controlled trials of CKD and dialysis cohorts

Study Duration Sample Size (n); CKD Diet/Phosphate Intake Binder/Dose FGF23 Assay FGF23 and Mineral Response
Binder versus placebo
 Block et al. (99) 9 mo 148; CKD stages 3 and 4; serum P=3.5–6.0 mg/dl Not controlled Composite active group: LaCO3, CaAc, SCO3; mean=2700, 5900, 6300 mg/d, respectively cFGF23/iFGF23 cFGF23/iFGF23: no change in active versus placebo; cFGF23: no difference between binder types; iFGF23: decrease in patients on SCO3, increase in patients on CaAc; modest decrease in serum P (4.2–3.9 mg/dl) and FEP (32%–26%) in active arm
 Isakova et al. (91) 3 mo (2×2) 39; CKD stages 3 and 4 normophosphatemic Ad libitum versus 900 mg LaCO3: fixed 3000 mg/d cFGF23 Neither LaCO3 nor 900 mg P diet alone reduced levels compared with placebo or ad libitum diet; variable reduction 35% (±32%) with dual LaCO3 and 900 mg P but baseline cFGF23 much higher in this group (mean=514 versus 99, 127, 120 RU/ml); no change in serum P or FEP
 Chue et al. (107) 9 mo 109; CKD stage 3 normophosphatemic Not controlled LaCO3: fixed 1600 mg/d iFGF23 No change on active therapy (median decrease=5 pg/ml); no change in serum P or FEP
 Seifert et al. (144) 12 mo 38; CKD stage 3 normophosphatemic Not controlled LaCO3: fixed 3000 mg/d iFGF23 No change in LaCO3 versus placebo; no change in serum or 24-h urine P
Calcium-based binder versus noncalcium-based binder
 CKD
  Oliveira et al. (145) 1.5 mo 40; CKD stages 3 and 4 normophosphatemic Dietary monitoring; 739±253 mg/d CaAc versus SHCl; titrated, doubled every 2 wk iFGF23 No change with CaAc (although trend to reduction); reduction by week 6 with SHCl (median decrease=49 pg/ml, 48%).; no change in serum P but decrease in FEP in both groups
  Yilmaz et al. (105) 2 mo 100; CKD stage 4 hyperphosphatemic Not controlled CaAc versus SHCl; titrated to serum P<5.5 mg/dl iFGF23 No change with CaAc (mean increase=4%); mean 27% reduction with SHCl (−33% to −9%); greater reduction in serum P with SHCl (mean decrease=31% versus 15%)
  Vlassara et al. (106) 2 wk (crossover) 22; CKD stages 2–4; diabetic Dietary monitoring; 967±479 mg/d CaCO3 versus SCO3; fixed 3600 versus 4800 mg/d iFGF23 Nonsignificant trend to reduction with SCO3 and increase with CaCO3; significant decrease with SCO3 treatment in patients with baseline concentration>70 pg/ml; no change in serum P but decrease in 24-h urine P
  Soriano et al. (146) 4 mo 32; CKD stages 4 and 5 hyperphosphatemic Standardized diet: 1 g/kg per day protein; <800 mg P CaCO3 versus LaCO3; titrated to serum P (<4.5 mg/dl) iFGF23 No reduction with CaCO3 but significant decrease with LaCO3 (mean decrease=36%); no change in serum P but decrease in FEP in LaCO3 arm
 Hemodialysis
  Koiwa et al. (147) 1 mo 46; prevalent HD Not controlled CaCO3 versus SHCl+CaCO3; fixed 3000 mg/d CaCO3 versus 3000 mg/d SHCl+3000 mg/d CaCO3 iFGF23 Reduction with CaCO3+SHCl but not with CaCO3 alone; levels increased in 8 of 26 patients on SHCl+CaCO3 therapy
  Cancela et al. (148) 12 mo 71; prevalent HD Not controlled CaAc versus SHCl; titrated to achieve serum P, iCal, iPTH targets iFGF23 Substantial reduction in patients receiving SHCl over 1 yr (median decrease=17,508–4627 pg/ml) but no change in patients on CaAc
  Toida et al. (149) 3 mo (crossover) 42; prevalent HD Not controlled CaCO3 versus LaCO3; titrated to achieve serum P, iCal, iPTH targets iFGF23 No change with CaCO3 but reduction with LaCO3 (mean decrease=46%)

P, phosphate; LaCO3, lanthanum carbonate; CaAc, calcium acetate; SCO3, sevelamer carbonate; SHCl, sevelamer hydrochloride; CaCO3, calcium carbonate; HD, hemodialysis; iCal, ionized calcium; iPTH, intact PTH.

The only tentative evidence of benefit associated with FGF23 lowering with an oral phosphate binder in the predialysis setting comes from a randomized, open-label study by Yilmaz et al. (105). An 8-week intervention with sevelamer hydrochloride in 47 hyperphosphatemic stage 4 CKD patients was associated with a 27% reduction in iFGF23 and a modest improvement in flow-mediated vasodilatation, independent of changes in serum phosphate. However, such an effect has yet to be replicated in other studies and remains mechanistically ambiguous: it may, in part, relate to the known pleiotropic effects of the drug (e.g., anti-inflammatory) (106). Other similarly sized trials in normophosphatemic CKD patients have failed to show efficacy in FGF23 lowering or in the case of the Chronic Renal Impairment in Birmingham-Phosphate study (107), improvement in outcome (LVMI) over 9 months of therapy.

On theoretical grounds, one might challenge the notion that phosphate modulation would significantly alter FGF23 levels given that phosphate, unlike 1,25(OH)2D or PTH, seems to have no direct effect on FGF23 production in cultured osteoblasts (2). Likewise, in healthy adults, intravenous infusion of phosphate fails to stimulate a response in FGF23 (108), and there is minimal acute postprandial change in FGF23 in response to dietary intake (23). Thus, any effect of phosphate on FGF23 would be modest (if existent at all) and sluggish, possibly caused by indirect and delayed effects on bone mineralization or mediated by secondary changes in 1,25(OH)2D or PTH. Regardless, if the desired effect of dietary phosphate restriction is to lower FGF23 concentrations, then the lack of a homogeneous and reproducible effect casts doubt on its clinical efficacy as a solitary intervention.

Vitamin D

Expectedly, given the stimulating effect of 1,25(OH)2D on FGF23 transcription (2), use of active vitamin D or its analogs (paracalcitol and alphacalcitol) has been associated with increased plasma FGF23 concentrations in patients with secondary hyperparathyroidism (109,110). The long-term consequence of this increase with respect to cardiovascular risk has not been explored, but strong evidence relating higher FGF23 with worse outcome possibly cautions against excessive active vitamin D use. Indeed, although observational studies of hemodialysis patients suggest a survival advantage with active vitamin D therapy (111) (albeit attenuated at high doses) (112) and studies in non-CKD animal models attest to the protective properties of vitamin D receptor agonists (VDRAs) on cardiac hypertrophy (113,114), recent randomized controlled trials in CKD patients have failed to show any improvement in left ventricular structure or function with paracalcitol versus placebo (115,116). Thus, in the setting of CKD, the benefit of VDRAs on the heart seems uncertain. Regarding FGF23, however, the response to vitamin D therapy is highly variable and unrelated to pretreatment levels, and in a subset of individuals, the change seems negligible. Although it is not clear whether the efficacy of therapy might differ in such patients, it remains possible that those individuals showing minimal change in FGF23 might be those patients that benefit most from treatment with VDRAs.

With respect to nutritional vitamin D therapy, high-dose ergocalciferol (300,000 IU) in osteoporotic individuals with vitamin D insufficiency but without renal disease yields a substantial increase in plasma FGF23 concentrations that is secondary to increases in phosphate and 1,25(OH)2D (117). Lower-dose therapy (<50,000 IU) with either ergocalciferol or cholecalciferol, however, has not been associated with significant changes in FGF23 in either hemodialysis patients or healthy postmenopausal women (118,119).

An alternative but hypothetical possibility is that, as an inducer of α-klotho expression (120), vitamin D therapy may help to restore the native FGF23–klotho axis in the kidney and parathyroid glands and hence, diminish end organ resistance and feedback. Theoretically, it might reduce systemic levels and attenuate some of the toxic effects of FGF23 excess. Renal 1,25(OH)2D depletion may also potentiate α-klotho deficiency through activation of the renin-angiotensin system and generation of angiotensin II (121). Interestingly, several reports of renin-angiotensin system inhibition in rodent CKD models suggest that such interventions may have beneficial effects on the FGF23–klotho axis by restoring renal α-klotho expression (122) and augmenting phosphate excretion while moderating the increase in systemic FGF23 levels with disease progression (87). Likewise, in CKD mice, VDRAs were found to increase soluble klotho levels in the serum and urine (but not the kidney), increase phosphaturia, and lower serum FGF23 concentration (123). Currently, however, clinical data in support of this hypothesis are lacking.

Calcimimetics

Use of cinacalcet or experimental calcimimetic compounds (e.g., velcalcetide) has been associated with reductions in FGF23 concentration in dialysis patients with secondary hyperparathyroidism (124127). A similar reduction has also been observed in rats with CKD equivalent to stages 3 and 4 in humans (128). Given in vitro and in vivo data indicating a direct stimulating effect of PTH on FGF23 synthesis through PTH receptor-1 signaling pathways (129131), one might expect the lowering effect of cinacalcet on FGF23 to be mediated by changes in PTH. Surprisingly, in dialysis patients, the effect of cinacalcet on FGF23 seems to be independent of changes in PTH (124127) but instead, associated with changes in serum phosphate and calcium in most studies (124,126,127), although not consistently (125). Indeed, not all studies support a direct role for PTH in regulating FGF23 (132), and infusion of (1–34)PTH peptide in humans and mice has been shown to result in acute reductions in circulating FGF23 rather than the expected increase in levels (133,134). Thus, a detailed mechanistic appraisal of how cinacalcet lowers FGF23 levels and the role of PTH [either directly or indirectly through changes in 1,25(OH)2D and/or local processes in bone] (133,135,136) is certainly needed. The response in FGF23 levels to cinacalcet therapy is, like for PTH, quite variable: approximately one half of patients show little (if any) change in concentrations in FGF23 (124,126), and responsiveness seems unrelated to pretreatment levels (137).

Despite the disappointment of recent data from the Evaluation of Cinacalcet Hydrochloride Therapy to Lower Cardiovascular Events trial (138), yielding a negative primary end point (a composite of all-cause mortality and nonfatal cardiovascular events), the biochemical profile associated with cinacalcet therapy seems favorable compared with active vitamin D therapy. Whether reductions in FGF23 with cinacalcet can be related to improvement in specific outcome measures (e.g., LVH) has yet to be reported. Theoretically, concurrent use of cinacalcet with a low-dose VDRA may be particularly useful in advanced CKD, combining the potentially beneficial cardiovascular effects of activation of the vitamin D receptor while lowering FGF23 concentrations. Again, however, such a positive effect has yet to be demonstrated in the clinical arena.

Experimental Approaches—FGF23 Neutralization and FGF Receptor Blockade

Administration of neutralizing FGF23 antibodies to 5/6-nephrectomized rats (with renal function equivalent to CKD stages 2–4 in humans) fed a high-phosphate diet, although normalizing calcium, 1,25(OH)2D, and PTH and improving some skeletal mineralization and volume parameters, results in profound hyperphosphatemia, extensive vascular calcifications, and reduced survival (57). Pan-FGF receptor inhibition in rats also gives rise to a tumoral calcinosis-like phenotype (139). Thus, in patients with nondialysis-dependent CKD, complete blockade of FGF23 action would seem undesirable. Whether neutralization in patients with ESRD, in whom FGF23 concentrations are often substantially higher and there is a plausible mechanism of direct FGF23 toxicity to the heart, would retard LVH progression and yield clinical benefit remains questionable but may provide a rationale for testing such interventions in this setting. Partial antagonism of circulating FGF23 or more selective inhibition of specific FGF receptor isoforms that block harmful effects on the heart (but permit signaling in the kidney and modulation of phosphate handling) might be more widely applicable but have yet to be tested.

Summary and Conclusions

Table 5 appraises how FGF23 performs alongside other bone mineral markers with respect to important analytical and clinical benchmarks using current state of the art methodology. Several characteristics may favor the use of FGF23 in CKD over conventional mineral markers, like PTH and phosphate: (1) temporal stability of the signal, (2) temporality of increase, and (3) greater discriminative power. Critically, unlike most other bone mineral markers, higher concentrations seem robustly associated with hard outcome measures, yielding large and stable effect sizes. Moreover, unlike other mineral metabolites, FGF23 seems reproducibly related to specific clinical pathologic correlates (e.g., LVH), which provides a strong rationale for the assessment of interventions aimed at lowering circulating FGF23 levels. In general, however, conventional CKD-MBD therapies (e.g., oral phosphate binders or calcimimetics) result in variable and unpredictable changes in FGF23 levels, similar to the changes in other markers of mineral metabolism. Moreover, mechanistic aspects of how FGF23 lowering is achieved with these therapies remain poorly defined and need to be delineated. Additional specific strategies for controlling FGF23 should be considered.

Table 5.

Performance of FGF23 and other systemic bone mineral markers against key analytical and clinical benchmarks

Benchmark Calcium Phosphate 25(OH)D 1,25(OH)2D PTH FGF23
Analytical criteria
 Biologic stability (diurnal, postprandial, week to week, interracial) Small magnitude diurnal variation and postprandial changes (23); minimal week-to-week variability (24,25,151) Substantial diurnal and week-to-week variability (15%–35%) (24,150,152,153) and marked postprandial and postexercise excursions (23) Marked seasonal (>100%) (26) and interracial variation (lower in African Americans than whites) (154,155) Modest diurnal and week-to-week variability (26,156) Very short plasma half-life (<4 min) (157), substantial diurnal (bimodal) and week-to-week variability (20%–40%) (153,158), greater variability in dialysis patients (25), significant interracial differences (higher in African Americans) (154,155) iFGF23: short plasma half-life (<60 min) (159), significant diurnal (30%) and week-to-week variability (20%) (20,22); cFGF23: minimal intraindividual variability (20,39), minimal postprandial response (23,91); significant interracial differences (lower in African Americans than whites) (46,154)
 Preanalytical issues Total: venous stasis/hemoconcentration effects; free: pH and temperature dependence Sample type differences; artifactual rise with delayed separation and hemolysis Negligible Negligible Sample type-, method-, and time-dependent stability (160,161) Sample type-, method-, and time-dependent stability (11,13)
 Measure and homogeneity Good Good Total D2+D3 or D3-specific methods; vitamin D binding protein bound and unbound fractions (bioavailable) (162) Poorly characterized protein binding profile Multiple fragments, method-dependent detection (32); partially characterized C-terminal fragments variably present (20); poorly characterized
 Between-method agreement Good Good Poor; calibration differences; matrix effects caused by serum binding proteins (164); variable cross-reactivity with other hydroxylated vitamin D metabolites (33); isobaric interferences in MS methods (33) Uncharacterized Poor (165,166); calibration differences; no reference method; variable antibody specificity and affinities for different PTH peptides (167); cross-reactivity of intact assays for PTH (784)—overestimates bioactive fraction (168) Poor; calibration differences (11,12); no reference method or ISP; variable fragment detection (20)
 High-throughput testing Yes Yes Yes No Yes No
 Cost per test (US)a $8 $8 $30 $40 $30 $40
Clinical criteria
 Temporality of change in CKD (3,3436) Variable CKD stages 4 and 5 Variable CKD stages 2 and 3 CKD stages 3 and 4 CKD stages 2 and 3
 Robust and reproducible independent association with hard outcomes in multiple CKD studiesb No (169) Multiple studies link higher P to increased risk for CV disease and mortality (169171) but not consistently (172) (Table 3) Several studies report association of low levels with and mortality (173), although not consistently (Table 3); few assessing outcome with bioavailable fraction Inconsistent in the small number of available studies, infrequently measured (Table 3) Inconsistent (169) (Table 3); few studies assessing outcome with whole/bioactive evels Consistent linear association with CV events, mortality, and progression to ESRD in multiple large prospective cohort studies; minimally confounded; substantial adjusted effect size (Table 2)
 Specific clinicopathological correlates Uncertain Vascular toxin (174)? (vascular calcification; endothelial dysfunction); LVH (175)? Tubular toxicity (163)? Deficiency linked to glomerular dysfunction (podocyte loss, glomerulosclerosis, albuminuria) and LVH (176)? Uncertain Variable association with bone histomorphometry (bone formation rate) except at extreme values (32) LVMI and LVH (7,6165)
 Adds to or superior performance over existing testsc Potential risk modifier (36) Potential risk modifier (54,56) Yes, improves discrimination and reclassification of risk associated with CV events and renal failure (42,45,47)
 Validated decision limits in CKD None
 Modifiable risk in CKD Uncertain
Variable response to therapy
Need to individualize therapy
 Marker-guided therapy improves outcome in CKD No evidence

MS, mass spectrometry; ISP, International Standard Preparation; CV, cardiovascular; LVH, left ventricular hypertrophy; LVMI, left ventricular mass index.

a

Approximate indicative commercial cost (including overheads).

b

Death or progression to ESRD requiring RRT.

c

Exisiting tests: calcium, phosphate, and PTH.

Seemingly, one of the key obstacles to incorporating FGF23 measurement into current clinical practice is the lack of evidence that lowering FGF23 results in benefit to the patient. However, equivalent data showing improvement in outcome associated with modulating other contemporary mineral measures (e.g., phosphate and PTH) are also lacking. For instance, in CKD patients, there remains no definitive evidence that serum phosphate lowering with oral binder therapy is associated with improvement in survival or for that matter, that restricting intestinal phosphate absorption even has the intended effect on overall phosphate balance. Indeed, in a recent study of stages 3 and 4 CKD patients treated with oral calcium carbonate, binder use was found to have no net effect on phosphate balance, at least in the short term (137). Thus, it could be argued that needing evidence of attenuated risk with FGF23 lowering demands substantially more from FGF23 than we ask of the mineral markers currently in use. Nonetheless, other questions regarding the use of FGF23 measurements have yet to be addressed. What is the goal of FGF23-directed therapy? Should it be to normalize FGF23 levels or restore a functional FGF23–klotho axis? Should we aim to neutralize the action of FGF23 or treat the underlying factors leading to its increase? What magnitude of reduction in FGF23 is required for a significant benefit? Given the substantial economic and practical implications of integrating a new biomarker into CKD management protocols, it seems prudent to rigorously assess the credentials of FGF23 measurement and explicitly define its clinical value in addition to or as an alternative to contemporary testing strategies. Indeed, perhaps it is timely to revisit the evidence base (or lack thereof) supporting the use of other routinely and frequently measured bone mineral markers. Assuming that FGF23 is toxic at high concentrations and that interventions to lower FGF23 yield clinical benefit, a number of potential applications for FGF23 measurement can be envisaged (Table 6).

Table 6.

Potential applications of FGF23 measurement in clinical nephrology

Application Example
Diagnostic Early biochemical detection of the CKD-MBD; renal dysfunction (e.g., tubulointerstitial injury)
Risk stratification CV: cardiac dysfunction, LVH, and death; high risk: high FGF23/low FEP/low 1,25(OH)2D; renal: rapid progressors (eGFR decline>3–5 ml/min per year), ESRD, and requirement for RRT; high risk: high FGF23/low 25(OH)D
Therapeutic selectiona Early CKD: noncalcium-based oral P binders plus low P dietary prescription; vitamin D therapy; ACEi/ARB; advanced CKD: calcimimetic use or concurrent calcimimetic plus low-dose VDRA; postrenal transplantation; P/calcitriol supplementation
Monitor therapeutic response As above; limit VDRA dose

CKD-MBD, CKD–mineral bone disorder; CV, cardiovascular; P, phosphate; ACEi/ARB, angiotensin-converting enzyme inhibitor/angiotensin II receptor blocker; VDRA, vitamin D receptor activator.

a

In addition to standard indications.

In aggregate, although FGF23 shows considerable promise as a biochemical target for guiding patient management and therapy, several gaps in our knowledge provide a barrier to the use of FGF23 testing in the clinic. Chief among these gaps are the lack of evidence linking FGF23 lowering with improved outcome and to some extent, the lack of a proven therapy to specifically and effectively target FGF23 excess. Fundamentally, however, we have yet to fully understand the mechanisms driving FGF23 elevations in CKD or the means by which therapies, like calcimimetics or phosphate restriction, act to lower circulating levels in some patients. Although a better mechanistic understanding of these pathways may also help reveal alternative or improved avenues for therapeutic intervention, such considerations seem essential to validate FGF23 as a genuine clinical target.

Disclosures

E.R.S. has received research funding from Amgen and Baxter, received honoraria from Shire, and served as a consultant for Vifor Pharma.

Footnotes

Published online ahead of print. Publication date available at www.cjasn.org.

References

  • 1.Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, Fukumoto S, Tomizuka K, Yamashita T: Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest 113: 561–568, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Liu S, Tang W, Zhou J, Stubbs JR, Luo Q, Pi M, Quarles LD: Fibroblast growth factor 23 is a counter-regulatory phosphaturic hormone for vitamin D. J Am Soc Nephrol 17: 1305–1315, 2006 [DOI] [PubMed] [Google Scholar]
  • 3.Isakova T, Wahl P, Vargas GS, Gutiérrez OM, Scialla J, Xie H, Appleby D, Nessel L, Bellovich K, Chen J, Hamm L, Gadegbeku C, Horwitz E, Townsend RR, Anderson CA, Lash JP, Hsu CY, Leonard MB, Wolf M: Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease. Kidney Int 79: 1370–1378, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Isakova T, Xie H, Yang W, Xie D, Anderson AH, Scialla J, Wahl P, Gutiérrez OM, Steigerwalt S, He J, Schwartz S, Lo J, Ojo A, Sondheimer J, Hsu CY, Lash J, Leonard M, Kusek JW, Feldman HI, Wolf M, Chronic Renal Insufficiency Cohort (CRIC) Study Group : Fibroblast growth factor 23 and risks of mortality and end-stage renal disease in patients with chronic kidney disease. JAMA 305: 2432–2439, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kendrick J, Cheung AK, Kaufman JS, Greene T, Roberts WL, Smits G, Chonchol M, HOST Investigators : FGF-23 associates with death, cardiovascular events, and initiation of chronic dialysis. J Am Soc Nephrol 22: 1913–1922, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Scialla JJ, Xie H, Rahman M, Anderson AH, Isakova T, Ojo A, Zhang X, Nessel L, Hamano T, Grunwald JE, Raj DS, Yang W, He J, Lash JP, Go AS, Kusek JW, Feldman H, Wolf M, the Chronic Renal Insufficiency Cohort (CRIC) Study Investigators : Fibroblast growth factor-23 and cardiovascular events in CKD [published online ahead of print October 24, 2013]. J Am Soc Nephrol 10.1681/ASN.2013050465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, Gutiérrez OM, Aguillon-Prada R, Lincoln J, Hare JM, Mundel P, Morales A, Scialla J, Fischer M, Soliman EZ, Chen J, Go AS, Rosas SE, Nessel L, Townsend RR, Feldman HI, St John Sutton M, Ojo A, Gadegbeku C, Di Marco GS, Reuter S, Kentrup D, Tiemann K, Brand M, Hill JA, Moe OW, Kuro-O M, Kusek JW, Keane MG, Wolf M: FGF23 induces left ventricular hypertrophy. J Clin Invest 121: 4393–4408, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.White KE, Carn G, Lorenz-Depiereux B, Benet-Pages A, Strom TM, Econs MJ: Autosomal-dominant hypophosphatemic rickets (ADHR) mutations stabilize FGF-23. Kidney Int 60: 2079–2086, 2001 [DOI] [PubMed] [Google Scholar]
  • 9.Benet-Pagès A, Lorenz-Depiereux B, Zischka H, White KE, Econs MJ, Strom TM: FGF23 is processed by proprotein convertases but not by PHEX. Bone 35: 455–462, 2004 [DOI] [PubMed] [Google Scholar]
  • 10.Shimada T, Muto T, Urakawa I, Yoneya T, Yamazaki Y, Okawa K, Takeuchi Y, Fujita T, Fukumoto S, Yamashita T: Mutant FGF-23 responsible for autosomal dominant hypophosphatemic rickets is resistant to proteolytic cleavage and causes hypophosphatemia in vivo. Endocrinology 143: 3179–3182, 2002 [DOI] [PubMed] [Google Scholar]
  • 11.Smith ER, McMahon LP, Holt SG: Method-specific differences in plasma fibroblast growth factor 23 measurement using four commercial ELISAs. Clin Chem Lab Med 51: 1971–1981, 2013 [DOI] [PubMed] [Google Scholar]
  • 12.Sinha MD, Turner C, Goldsmith DJ: FGF23 concentrations measured using “intact” assays similar but not interchangeable. Int Urol Nephrol 45: 1821–1823, 2013 [DOI] [PubMed] [Google Scholar]
  • 13.Smith ER, Ford ML, Tomlinson LA, Weaving G, Rocks BF, Rajkumar C, Holt SG: Instability of fibroblast growth factor-23 (FGF-23): Implications for clinical studies. Clin Chim Acta 412: 1008–1011, 2011 [DOI] [PubMed] [Google Scholar]
  • 14.Jonsson KB, Zahradnik R, Larsson T, White KE, Sugimoto T, Imanishi Y, Yamamoto T, Hampson G, Koshiyama H, Ljunggren O, Oba K, Yang IM, Miyauchi A, Econs MJ, Lavigne J, Jüppner H: Fibroblast growth factor 23 in oncogenic osteomalacia and X-linked hypophosphatemia. N Engl J Med 348: 1656–1663, 2003 [DOI] [PubMed] [Google Scholar]
  • 15.Wolf M, Koch TA, Bregman DB: Effects of iron deficiency anemia and its treatment on fibroblast growth factor 23 and phosphate homeostasis in women. J Bone Miner Res 28: 1793–1803, 2013 [DOI] [PubMed] [Google Scholar]
  • 16.Bhattacharyya N, Wiench M, Dumitrescu C, Connolly BM, Bugge TH, Patel HV, Gafni RI, Cherman N, Cho M, Hager GL, Collins MT: Mechanism of FGF23 processing in fibrous dysplasia. J Bone Miner Res 27: 1132–1141, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Larsson T, Davis SI, Garringer HJ, Mooney SD, Draman MS, Cullen MJ, White KE: Fibroblast growth factor-23 mutants causing familial tumoral calcinosis are differentially processed. Endocrinology 146: 3883–3891, 2005 [DOI] [PubMed] [Google Scholar]
  • 18.Berndt TJ, Craig TA, McCormick DJ, Lanske B, Sitara D, Razzaque MS, Pragnell M, Bowe AE, O’Brien SP, Schiavi SC, Kumar R: Biological activity of FGF-23 fragments. Pflugers Arch 454: 615–623, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Goetz R, Nakada Y, Hu MC, Kurosu H, Wang L, Nakatani T, Shi M, Eliseenkova AV, Razzaque MS, Moe OW, Kuro-o M, Mohammadi M: Isolated C-terminal tail of FGF23 alleviates hypophosphatemia by inhibiting FGF23-FGFR-Klotho complex formation. Proc Natl Acad Sci U S A 107: 407–412, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Smith ER, Cai MM, McMahon LP, Holt SG: Biological variability of plasma intact and C-terminal FGF23 measurements. J Clin Endocrinol Metab 97: 3357–3365, 2012 [DOI] [PubMed] [Google Scholar]
  • 21.Fischer DC, Mischek A, Wolf S, Rahn A, Salweski B, Kundt G, Haffner D: Paediatric reference values for the C-terminal fragment of fibroblast-growth factor-23, sclerostin, bone-specific alkaline phosphatase and isoform 5b of tartrate-resistant acid phosphatase. Ann Clin Biochem 49: 546–553, 2012 [DOI] [PubMed] [Google Scholar]
  • 22.Vervloet MG, van Ittersum FJ, Büttler RM, Heijboer AC, Blankenstein MA, ter Wee PM: Effects of dietary phosphate and calcium intake on fibroblast growth factor-23. Clin J Am Soc Nephrol 6: 383–389, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Isakova T, Gutierrez O, Shah A, Castaldo L, Holmes J, Lee H, Wolf M: Postprandial mineral metabolism and secondary hyperparathyroidism in early CKD. J Am Soc Nephrol 19: 615–623, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pocock SJ, Ashby D, Shaper AG, Walker M, Broughton PM: Diurnal variations in serum biochemical and haematological measurements. J Clin Pathol 42: 172–179, 1989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gardham C, Stevens PE, Delaney MP, LeRoux M, Coleman A, Lamb EJ: Variability of parathyroid hormone and other markers of bone mineral metabolism in patients receiving hemodialysis. Clin J Am Soc Nephrol 5: 1261–1267, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hine TJ, Roberts NB: Seasonal variation in serum 25-hydroxy vitamin D3 does not affect 1,25-dihydroxy vitamin D. Ann Clin Biochem 31: 31–34, 1994 [DOI] [PubMed] [Google Scholar]
  • 27.Fraser CG, Harris EK: Generation and application of data on biological variation in clinical chemistry. Crit Rev Clin Lab Sci 27: 409–437, 1989 [DOI] [PubMed] [Google Scholar]
  • 28.Seiler S, Lucisano G, Ege P, Fell LH, Rogacev KS, Lerner-Gräber A, Klingele M, Ziegler M, Fliser D, Heine GH: Single FGF-23 measurement and time-averaged plasma phosphate levels in hemodialysis patients. Clin J Am Soc Nephrol 8: 1764–1772, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cavalier E, Delanaye P, Moranne O: Variability of new bone mineral metabolism markers in patients treated with maintenance hemodialysis: Implications for clinical decision making. Am J Kidney Dis 61: 847–848, 2013 [DOI] [PubMed] [Google Scholar]
  • 30.Jia T, Qureshi AR, Brandenburg V, Ketteler M, Barany P, Heimburger O, Uhlin F, Magnusson P, Fernström A, Lindholm B, Stenvinkel P, Larsson TE: Determinants of fibroblast growth factor-23 and parathyroid hormone variability in dialysis patients. Am J Nephrol 37: 462–471, 2013 [DOI] [PubMed] [Google Scholar]
  • 31.Shimizu Y, Fukumoto S, Fujita T: Evaluation of a new automated chemiluminescence immunoassay for FGF23. J Bone Miner Metab 30: 217–221, 2012 [DOI] [PubMed] [Google Scholar]
  • 32.Garrett G, Sardiwal S, Lamb EJ, Goldsmith DJ: PTH—a particularly tricky hormone: Why measure it at all in kidney patients? Clin J Am Soc Nephrol 8: 299–312, 2013 [DOI] [PubMed] [Google Scholar]
  • 33.Carter GD: 25-hydroxyvitamin D: A difficult analyte. Clin Chem 58: 486–488, 2012 [DOI] [PubMed] [Google Scholar]
  • 34.Levin A, Bakris GL, Molitch M, Smulders M, Tian J, Williams LA, Andress DL: Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: Results of the study to evaluate early kidney disease. Kidney Int 71: 31–38, 2007 [DOI] [PubMed] [Google Scholar]
  • 35.Portale AA, Wolf M, Juppner H, Messinger S, Kumar J, Wesseling-Perry K, Schwartz GJ, Furth SL, Warady BA, Salusky IB: Disordered FGF23 and mineral metabolism in children with CKD [published online ahead of print December 5, 2013]. Clin J Am Soc Nephrol 10.2215/CJN.05840513 [DOI] [PMC free article] [PubMed]
  • 36.Nakano C, Hamano T, Fujii N, Matsui I, Tomida K, Mikami S, Inoue K, Obi Y, Okada N, Tsubakihara Y, Isaka Y, Rakugi H: Combined use of vitamin D status and FGF23 for risk stratification of renal outcome. Clin J Am Soc Nephrol 7: 810–819, 2012 [DOI] [PubMed] [Google Scholar]
  • 37.Stubbs JR, He N, Idiculla A, Gillihan R, Liu S, David V, Hong Y, Quarles LD: Longitudinal evaluation of FGF23 changes and mineral metabolism abnormalities in a mouse model of chronic kidney disease. J Bone Miner Res 27: 38–46, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Shimada T, Urakawa I, Isakova T, Yamazaki Y, Epstein M, Wesseling-Perry K, Wolf M, Salusky IB, Jüppner H: Circulating fibroblast growth factor 23 in patients with end-stage renal disease treated by peritoneal dialysis is intact and biologically active. J Clin Endocrinol Metab 95: 578–585, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Isakova T, Xie H, Barchi-Chung A, Vargas G, Sowden N, Houston J, Wahl P, Lundquist A, Epstein M, Smith K, Contreras G, Ortega L, Lenz O, Briones P, Egbert P, Ikizler TA, Jueppner H, Wolf M: Fibroblast growth factor 23 in patients undergoing peritoneal dialysis. Clin J Am Soc Nephrol 6: 2688–2695, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wesseling-Perry K, Pereira RC, Wang H, Elashoff RM, Sahney S, Gales B, Jüppner H, Salusky IB: Relationship between plasma fibroblast growth factor-23 concentration and bone mineralization in children with renal failure on peritoneal dialysis. J Clin Endocrinol Metab 94: 511–517, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Pande S, Ritter CS, Rothstein M, Wiesen K, Vassiliadis J, Kumar R, Schiavi SC, Slatapolsky E, Brown AJ: FGF-23 and sFRP-4 in chronic kidney disease and post-renal transplantation. Nephron Physiol 104: 23–32, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Nakano C, Hamano T, Fujii N, Obi Y, Matsui I, Tomida K, Mikami S, Inoue K, Shimomura A, Nagasawa Y, Okada N, Tsubakihara Y, Rakugi H, Isaka Y: Intact fibroblast growth factor 23 levels predict incident cardiovascular event before but not after the start of dialysis. Bone 50: 1266–1274, 2012 [DOI] [PubMed] [Google Scholar]
  • 43.Gutiérrez OM, Mannstadt M, Isakova T, Rauh-Hain JA, Tamez H, Shah A, Smith K, Lee H, Thadhani R, Jüppner H, Wolf M: Fibroblast growth factor 23 and mortality among patients undergoing hemodialysis. N Engl J Med 359: 584–592, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wolf M, Molnar MZ, Amaral AP, Czira ME, Rudas A, Ujszaszi A, Kiss I, Rosivall L, Kosa J, Lakatos P, Kovesdy CP, Mucsi I: Elevated fibroblast growth factor 23 is a risk factor for kidney transplant loss and mortality. J Am Soc Nephrol 22: 956–966, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Baia LC, Humalda JK, Vervloet MG, Navis G, Bakker SJ, de Borst MH, NIGRAM Consortium : Fibroblast growth factor 23 and cardiovascular mortality after kidney transplantation. Clin J Am Soc Nephrol 8: 1968–1978, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Scialla JJ, Astor BC, Isakova T, Xie H, Appel LJ, Wolf M: Mineral metabolites and CKD progression in African Americans. J Am Soc Nephrol 24: 125–135, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Smith ER, Ford ML, Tomlinson LA, McMahon LP, Rajkumar C, Holt SG: FGF23 adds value to risk prediction in patients with chronic kidney disease. Bone 51: 830–831, 2012 [DOI] [PubMed] [Google Scholar]
  • 48.Lundberg S, Qureshi AR, Olivecrona S, Gunnarsson I, Jacobson SH, Larsson TE: FGF23, albuminuria, and disease progression in patients with chronic IgA nephropathy. Clin J Am Soc Nephrol 7: 727–734, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Titan SM, Zatz R, Graciolli FG, dos Reis LM, Barros RT, Jorgetti V, Moysés RM: FGF-23 as a predictor of renal outcome in diabetic nephropathy. Clin J Am Soc Nephrol 6: 241–247, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Semba RD, Fink JC, Sun K, Cappola AR, Dalal M, Crasto C, Ferrucci L, Fried LP: Serum fibroblast growth factor-23 and risk of incident chronic kidney disease in older community-dwelling women. Clin J Am Soc Nephrol 7: 85–91, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Leaf DE, Wolf M, Waikar SS, Chase H, Christov M, Cremers S, Stern L: FGF-23 levels in patients with AKI and risk of adverse outcomes. Clin J Am Soc Nephrol 7: 1217–1223, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Brown JR, Katz R, Ix JH, de Boer IH, Siscovick DS, Grams ME, Shlipak M, Sarnak MJ: Fibroblast growth factor-23 and the long-term risk of hospital-associated AKI among community-dwelling older individuals [published online ahead of print November 21, 2013]. Clin J Am Soc Nephrol 0.2215/CJN.05830513 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Weiner DE, Tighiouart H, Elsayed EF, Griffith JL, Salem DN, Levey AS, Sarnak MJ: The Framingham predictive instrument in chronic kidney disease. J Am Coll Cardiol 50: 217–224, 2007 [DOI] [PubMed] [Google Scholar]
  • 54.Ky B, Shults J, Keane MG, Sutton MS, Wolf M, Feldman HI, Reese PP, Anderson CA, Townsend RR, Deo R, Lo J, Gadegbeku C, Carlow D, Sulik MJ, Leonard MB, CRIC Study Investigators : FGF23 modifies the relationship between vitamin D and cardiac remodeling. Circ Heart Fail 6: 817–824, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Dominguez JR, Shlipak MG, Whooley MA, Ix JH: Fractional excretion of phosphorus modifies the association between fibroblast growth factor-23 and outcomes. J Am Soc Nephrol 24: 647–654, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kendrick J, Cheung AK, Kaufman JS, Greene T, Roberts WL, Smits G, Chonchol M, HOST (Homocysteinemia in Kidney and End Stage Renal Disease) Study Investigators : Associations of plasma 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D concentrations with death and progression to maintenance dialysis in patients with advanced kidney disease. Am J Kidney Dis 60: 567–575, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Shalhoub V, Shatzen EM, Ward SC, Davis J, Stevens J, Bi V, Renshaw L, Hawkins N, Wang W, Chen C, Tsai MM, Cattley RC, Wronski TJ, Xia X, Li X, Henley C, Eschenberg M, Richards WG: FGF23 neutralization improves chronic kidney disease-associated hyperparathyroidism yet increases mortality. J Clin Invest 122: 2543–2553, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gutierrez O, Isakova T, Rhee E, Shah A, Holmes J, Collerone G, Jüppner H, Wolf M: Fibroblast growth factor-23 mitigates hyperphosphatemia but accentuates calcitriol deficiency in chronic kidney disease. J Am Soc Nephrol 16: 2205–2215, 2005 [DOI] [PubMed] [Google Scholar]
  • 59.Parker BD, Schurgers LJ, Brandenburg VM, Christenson RH, Vermeer C, Ketteler M, Shlipak MG, Whooley MA, Ix JH: 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 152: 640–648, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ix JH, Katz R, Kestenbaum BR, de Boer IH, Chonchol M, Mukamal KJ, Rifkin D, Siscovick DS, Sarnak MJ, Shlipak MG: Fibroblast growth factor-23 and death, heart failure, and cardiovascular events in community-living individuals: CHS (Cardiovascular Health Study). J Am Coll Cardiol 60: 200–207, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Seiler S, Cremers B, Rebling NM, Hornof F, Jeken J, Kersting S, Steimle C, Ege P, Fehrenz M, Rogacev KS, Scheller B, Böhm M, Fliser D, Heine GH: The phosphatonin fibroblast growth factor 23 links calcium-phosphate metabolism with left-ventricular dysfunction and atrial fibrillation. Eur Heart J 32: 2688–2696, 2011 [DOI] [PubMed] [Google Scholar]
  • 62.Seeherunvong W, Abitbol CL, Chandar J, Rusconi P, Zilleruelo GE, Freundlich M: Fibroblast growth factor 23 and left ventricular hypertrophy in children on dialysis. Pediatr Nephrol 27: 2129–2136, 2012 [DOI] [PubMed] [Google Scholar]
  • 63.Smith K, deFilippi C, Isakova T, Gutiérrez OM, Laliberte K, Seliger S, Kelley W, Duh SH, Hise M, Christenson R, Wolf M, Januzzi J: Fibroblast growth factor 23, high-sensitivity cardiac troponin, and left ventricular hypertrophy in CKD. Am J Kidney Dis 61: 67–73, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Gutiérrez OM, Januzzi JL, Isakova T, Laliberte K, Smith K, Collerone G, Sarwar A, Hoffmann U, Coglianese E, Christenson R, Wang TJ, deFilippi C, Wolf M: Fibroblast growth factor 23 and left ventricular hypertrophy in chronic kidney disease. Circulation 119: 2545–2552, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Negishi K, Kobayashi M, Ochiai I, Yamazaki Y, Hasegawa H, Yamashita T, Shimizu T, Kasama S, Kurabayashi M: Association between fibroblast growth factor 23 and left ventricular hypertrophy in maintenance hemodialysis patients. Comparison with B-type natriuretic peptide and cardiac troponin T. Circ J 74: 2734–2740, 2010 [DOI] [PubMed] [Google Scholar]
  • 66.Middleton RJ, Parfrey PS, Foley RN: Left ventricular hypertrophy in the renal patient. J Am Soc Nephrol 12: 1079–1084, 2001 [DOI] [PubMed] [Google Scholar]
  • 67.US Renal Data System : United States Renal Data System 2013 Annual Data Report: Atlas of Chronic Kidney Disease and End-Stage Renal Disease in the United States, Bethesda, MD, National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, 2013 [Google Scholar]
  • 68.Touchberry CD, Green TM, Tchikrizov V, Mannix JE, Mao TF, Carney BW, Girgis M, Vincent RJ, Wetmore LA, Dawn B, Bonewald LF, Stubbs JR, Wacker MJ: FGF23 is a novel regulator of intracellular calcium and cardiac contractility in addition to cardiac hypertrophy. Am J Physiol Endocrinol Metab 304: E863–E873, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, Fujita T, Fukumoto S, Yamashita T: Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature 444: 770–774, 2006 [DOI] [PubMed] [Google Scholar]
  • 70.Hu MC, Shi M, Zhang J, Quiñones H, Griffith C, Kuro-o M, Moe OW: Klotho deficiency causes vascular calcification in chronic kidney disease. J Am Soc Nephrol 22: 124–136, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Krajisnik T, Olauson H, Mirza MA, Hellman P, Akerström G, Westin G, Larsson TE, Björklund P: Parathyroid Klotho and FGF-receptor 1 expression decline with renal function in hyperparathyroid patients with chronic kidney disease and kidney transplant recipients. Kidney Int 78: 1024–1032, 2010 [DOI] [PubMed] [Google Scholar]
  • 72.Jovanovich A, Ix JH, Gottdiener J, McFann K, Katz R, Kestenbaum B, de Boer IH, Sarnak M, Shlipak MG, Mukamal KJ, Siscovick D, Chonchol M: Fibroblast growth factor 23, left ventricular mass, and left ventricular hypertrophy in community-dwelling older adults. Atherosclerosis 231: 114–119, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mirza MA, Larsson A, Melhus H, Lind L, Larsson TE: Serum intact FGF23 associate with left ventricular mass, hypertrophy and geometry in an elderly population. Atherosclerosis 207: 546–551, 2009 [DOI] [PubMed] [Google Scholar]
  • 74.Ärnlöv J, Carlsson AC, Sundström J, Ingelsson E, Larsson A, Lind L, Larsson TE: Serum FGF23 and risk of cardiovascular events in relation to mineral metabolism and cardiovascular pathology. Clin J Am Soc Nephrol 8: 781–786, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ärnlöv J, Carlsson AC, Sundström J, Ingelsson E, Larsson A, Lind L, Larsson TE: Higher fibroblast growth factor-23 increases the risk of all-cause and cardiovascular mortality in the community. Kidney Int 83: 160–166, 2013 [DOI] [PubMed] [Google Scholar]
  • 76.Dai B, David V, Martin A, Huang J, Li H, Jiao Y, Gu W, Quarles LD: A comparative transcriptome analysis identifying FGF23 regulated genes in the kidney of a mouse CKD model. PLoS One 7: e44161, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Munoz Mendoza J, Isakova T, Ricardo AC, Xie H, Navaneethan SD, Anderson AH, Bazzano LA, Xie D, Kretzler M, Nessel L, Hamm LL, Negrea L, Leonard MB, Raj D, Wolf M, Chronic Renal Insufficiency Cohort : Fibroblast growth factor 23 and Inflammation in CKD. Clin J Am Soc Nephrol 7: 1155–1162, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Silver J, Naveh-Many T: FGF-23 and secondary hyperparathyroidism in chronic kidney disease. Nat Rev Nephrol 9: 641–649, 2013 [DOI] [PubMed] [Google Scholar]
  • 79.Pereira RC, Juppner H, Azucena-Serrano CE, Yadin O, Salusky IB, Wesseling-Perry K: Patterns of FGF-23, DMP1, and MEPE expression in patients with chronic kidney disease. Bone 45: 1161–1168, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Moe SM, Radcliffe JS, White KE, Gattone VH, 2nd, Seifert MF, Chen X, Aldridge B, Chen NX: The pathophysiology of early-stage chronic kidney disease-mineral bone disorder (CKD-MBD) and response to phosphate binders in the rat. J Bone Miner Res 26: 2672–2681, 2011 [DOI] [PubMed] [Google Scholar]
  • 81.Christov M, Waikar SS, Pereira RC, Havasi A, Leaf DE, Goltzman D, Pajevic PD, Wolf M, Jüppner H: Plasma FGF23 levels increase rapidly after acute kidney injury. Kidney Int 84: 776–785, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Yoshiko Y, Wang H, Minamizaki T, Ijuin C, Yamamoto R, Suemune S, Kozai K, Tanne K, Aubin JE, Maeda N: Mineralized tissue cells are a principal source of FGF23. Bone 40: 1565–1573, 2007 [DOI] [PubMed] [Google Scholar]
  • 83.Saji F, Shiizaki K, Shimada S, Okada T, Kunimoto K, Sakaguchi T, Hatamura I, Shigematsu T: Regulation of fibroblast growth factor 23 production in bone in uremic rats. Nephron, Physiol 111: 59–66, 2009 [DOI] [PubMed] [Google Scholar]
  • 84.Shimada T, Mizutani S, Muto T, Yoneya T, Hino R, Takeda S, Takeuchi Y, Fujita T, Fukumoto S, Yamashita T: Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc Natl Acad Sci U S A 98: 6500–6505, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Mirams M, Robinson BG, Mason RS, Nelson AE: Bone as a source of FGF23: Regulation by phosphate? Bone 35: 1192–1199, 2004 [DOI] [PubMed] [Google Scholar]
  • 86.ADHR Consortium : Autosomal dominant hypophosphataemic rickets is associated with mutations in FGF23. Nat Genet 26: 345–348, 2000 [DOI] [PubMed] [Google Scholar]
  • 87.Zanchi C, Locatelli M, Benigni A, Corna D, Tomasoni S, Rottoli D, Gaspari F, Remuzzi G, Zoja C: Renal expression of FGF23 in progressive renal disease of diabetes and the effect of ace inhibitor. PLoS One 8: e70775, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Spichtig D, Zhang H, Mohebbi N, Pavik I, Petzold K, Stange G, Saleh L, Edenhofer I, Segerer S, Biber J, Jaeger P, Serra AL, Wagner CA: Renal expression of FGF23 and peripheral resistance to elevated FGF23 in rodent models of polycystic kidney disease [published online ahead of print January 8, 2014]. Kidney Int 10.1038/ki.2013.526 [DOI] [PubMed] [Google Scholar]
  • 89.Wesseling-Perry K, Pereira RC, Tsai E, Ettenger R, Jüppner H, Salusky IB: FGF23 and mineral metabolism in the early post-renal transplantation period. Pediatr Nephrol 28: 2207–2215, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Moe SM, Zidehsarai MP, Chambers MA, Jackman LA, Radcliffe JS, Trevino LL, Donahue SE, Asplin JR: Vegetarian compared with meat dietary protein source and phosphorus homeostasis in chronic kidney disease. Clin J Am Soc Nephrol 6: 257–264, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Isakova T, Barchi-Chung A, Enfield G, Smith K, Vargas G, Houston J, Xie H, Wahl P, Schiavenato E, Dosch A, Gutiérrez OM, Diego J, Lenz O, Contreras G, Mendez A, Weiner RB, Wolf M: Effects of dietary phosphate restriction and phosphate binders on FGF23 levels in CKD. Clin J Am Soc Nephrol 8: 1009–1018, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Di Iorio B, Di Micco L, Torraca S, Sirico ML, Russo L, Pota A, Mirenghi F, Russo D: Acute effects of very-low-protein diet on FGF23 levels: A randomized study. Clin J Am Soc Nephrol 7: 581–587, 2012 [DOI] [PubMed] [Google Scholar]
  • 93.Gonzalez-Parra E, Gonzalez-Casaus ML, Galán A, Martinez-Calero A, Navas V, Rodriguez M, Ortiz A: Lanthanum carbonate reduces FGF23 in chronic kidney disease Stage 3 patients. Nephrol Dial Transplant 26: 2567–2571, 2011 [DOI] [PubMed] [Google Scholar]
  • 94.Sigrist M, Tang M, Beaulieu M, Espino-Hernandez G, Er L, Djurdjev O, Levin A: Responsiveness of FGF-23 and mineral metabolism to altered dietary phosphate intake in chronic kidney disease (CKD): Results of a randomized trial. Nephrol Dial Transplant 28: 161–169, 2013 [DOI] [PubMed] [Google Scholar]
  • 95.David V, Dai B, Martin A, Huang J, Han X, Quarles LD: Calcium regulates FGF-23 expression in bone. Endocrinology 154: 4469–4482, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Moe SM, Chen NX, Newman CL, Gattone VH, 2nd, Organ JM, Chen X, Allen MR: A comparison of calcium to zoledronic acid for improvement of cortical bone in an animal model of CKD [published online ahead of print September 3, 2013]. J Bone Miner Res 10.1002/jbmr.2089 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Brandenburg VM, Schlieper G, Heussen N, Holzmann S, Busch B, Evenepoel P, Vanholder R, Meijers B, Meert N, Fassbender WJ, Floege J, Jahnen-Dechent W, Ketteler M: Serological cardiovascular and mortality risk predictors in dialysis patients receiving sevelamer: A prospective study. Nephrol Dial Transplant 25: 2672–2679, 2010 [DOI] [PubMed] [Google Scholar]
  • 98.Spatz C, Roe K, Lehman E, Verma N: Effect of a non-calcium-based phosphate binder on fibroblast growth factor 23 in chronic kidney disease. Nephron Clin Pract 123: 61–66, 2013 [DOI] [PubMed] [Google Scholar]
  • 99.Block GA, Wheeler DC, Persky MS, Kestenbaum B, Ketteler M, Spiegel DM, Allison MA, Asplin J, Smits G, Hoofnagle AN, Kooienga L, Thadhani R, Mannstadt M, Wolf M, Chertow GM: Effects of phosphate binders in moderate CKD. J Am Soc Nephrol 23: 1407–1415, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Isakova T, Gutiérrez OM, Smith K, Epstein M, Keating LK, Jüppner H, Wolf M: Pilot study of dietary phosphorus restriction and phosphorus binders to target fibroblast growth factor 23 in patients with chronic kidney disease. Nephrol Dial Transplant 26: 584–591, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Palomino HL, Rifkin DE, Anderson C, Criqui MH, Whooley MA, Ix JH: 24-hour urine phosphorus excretion and mortality and cardiovascular events. Clin J Am Soc Nephrol 8: 1202–1210, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Gutierrez OM, Wolf M, Taylor EN: Fibroblast growth factor 23, cardiovascular disease risk factors, and phosphorus intake in the health professionals follow-up study. Clin J Am Soc Nephrol 6: 2871–2878, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Maizel J, Six I, Dupont S, Secq E, Dehedin B, Barreto FC, Benchitrit J, Poirot S, Slama M, Tribouilloy C, Choukroun G, Mazière JC, Drueke TB, Massy ZA: Effects of sevelamer treatment on cardiovascular abnormalities in mice with chronic renal failure. Kidney Int 84: 491–500, 2013 [DOI] [PubMed] [Google Scholar]
  • 104.Zhang S, Gillihan R, He N, Fields T, Liu S, Green T, Stubbs JR: Dietary phosphate restriction suppresses phosphaturia but does not prevent FGF23 elevation in a mouse model of chronic kidney disease. Kidney Int 84: 713–721, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Yilmaz MI, Sonmez A, Saglam M, Yaman H, Kilic S, Eyileten T, Caglar K, Oguz Y, Vural A, Yenicesu M, Mallamaci F, Zoccali C: Comparison of calcium acetate and sevelamer on vascular function and fibroblast growth factor 23 in CKD patients: A randomized clinical trial. Am J Kidney Dis 59: 177–185, 2012 [DOI] [PubMed] [Google Scholar]
  • 106.Vlassara H, Uribarri J, Cai W, Goodman S, Pyzik R, Post J, Grosjean F, Woodward M, Striker GE: Effects of sevelamer on HbA1c, inflammation, and advanced glycation end products in diabetic kidney disease. Clin J Am Soc Nephrol 7: 934–942, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Chue CD, Townend JN, Moody WE, Zehnder D, Wall NA, Harper L, Edwards NC, Steeds RP, Ferro CJ: Cardiovascular effects of sevelamer in stage 3 CKD. J Am Soc Nephrol 24: 842–852, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Ito N, Fukumoto S, Takeuchi Y, Takeda S, Suzuki H, Yamashita T, Fujita T: Effect of acute changes of serum phosphate on fibroblast growth factor (FGF)23 levels in humans. J Bone Miner Metab 25: 419–422, 2007 [DOI] [PubMed] [Google Scholar]
  • 109.Wesseling-Perry K, Pereira RC, Sahney S, Gales B, Wang HJ, Elashoff R, Jüppner H, Salusky IB: Calcitriol and doxercalciferol are equivalent in controlling bone turnover, suppressing parathyroid hormone, and increasing fibroblast growth factor-23 in secondary hyperparathyroidism. Kidney Int 79: 112–119, 2011 [DOI] [PubMed] [Google Scholar]
  • 110.Hansen D, Rasmussen K, Pedersen SM, Rasmussen LM, Brandi L: Changes in fibroblast growth factor 23 during treatment of secondary hyperparathyroidism with alfacalcidol or paricalcitol. Nephrol Dial Transplant 27: 2263–2269, 2012 [DOI] [PubMed] [Google Scholar]
  • 111.Teng M, Wolf M, Lowrie E, Ofsthun N, Lazarus JM, Thadhani R: Survival of patients undergoing hemodialysis with paricalcitol or calcitriol therapy. N Engl J Med 349: 446–456, 2003 [DOI] [PubMed] [Google Scholar]
  • 112.Kalantar-Zadeh K, Kuwae N, Regidor DL, Kovesdy CP, Kilpatrick RD, Shinaberger CS, McAllister CJ, Budoff MJ, Salusky IB, Kopple JD: Survival predictability of time-varying indicators of bone disease in maintenance hemodialysis patients. Kidney Int 70: 771–780, 2006 [DOI] [PubMed] [Google Scholar]
  • 113.Bae S, Yalamarti B, Ke Q, Choudhury S, Yu H, Karumanchi SA, Kroeger P, Thadhani R, Kang PM: Preventing progression of cardiac hypertrophy and development of heart failure by paricalcitol therapy in rats. Cardiovasc Res 91: 632–639, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kong J, Kim GH, Wei M, Sun T, Li G, Liu SQ, Li X, Bhan I, Zhao Q, Thadhani R, Li YC: Therapeutic effects of vitamin D analogs on cardiac hypertrophy in spontaneously hypertensive rats. Am J Pathol 177: 622–631, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Thadhani R, Appelbaum E, Pritchett Y, Chang Y, Wenger J, Tamez H, Bhan I, Agarwal R, Zoccali C, Wanner C, Lloyd-Jones D, Cannata J, Thompson BT, Andress D, Zhang W, Packham D, Singh B, Zehnder D, Shah A, Pachika A, Manning WJ, Solomon SD: Vitamin D therapy and cardiac structure and function in patients with chronic kidney disease: The PRIMO randomized controlled trial. JAMA 307: 674–684, 2012 [DOI] [PubMed] [Google Scholar]
  • 116.Wang AY, Fang F, Chan J, Wen YY, Qing S, Chan IH, Lo G, Lai KN, Lo WK, Lam CW, Yu CM: Effect of paricalcitol on left ventricular mass and function in CKD—The OPERA Trial. J Am Soc Nephrol 25: 175–186, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Turner C, Dalton N, Inaoui R, Fogelman I, Fraser WD, Hampson G: Effect of a 300 000-IU loading dose of ergocalciferol (Vitamin D2) on circulating 1,25(OH)2-vitamin D and fibroblast growth factor-23 (FGF-23) in vitamin D insufficiency. J Clin Endocrinol Metab 98: 550–556, 2013 [DOI] [PubMed] [Google Scholar]
  • 118.Gravesen E, Hofman-Bang J, Lewin E, Olgaard K: Ergocalciferol treatment and aspects of mineral homeostasis in patients with chronic kidney disease stage 4-5. Scand J Clin Lab Invest 73: 107–116, 2013 [DOI] [PubMed] [Google Scholar]
  • 119.Seibert E, Heine GH, Ulrich C, Seiler S, Köhler H, Girndt M: Influence of cholecalciferol supplementation in hemodialysis patients on monocyte subsets: A randomized, double-blind, placebo-controlled clinical trial. Nephron Clin Pract 123: 209–219, 2013 [DOI] [PubMed] [Google Scholar]
  • 120.Tsujikawa H, Kurotaki Y, Fujimori T, Fukuda K, Nabeshima Y: Klotho, a gene related to a syndrome resembling human premature aging, functions in a negative regulatory circuit of vitamin D endocrine system. Mol Endocrinol 17: 2393–2403, 2003 [DOI] [PubMed] [Google Scholar]
  • 121.Mitani H, Ishizaka N, Aizawa T, Ohno M, Usui S, Suzuki T, Amaki T, Mori I, Nakamura Y, Sato M, Nangaku M, Hirata Y, Nagai R: In vivo klotho gene transfer ameliorates angiotensin II-induced renal damage. Hypertension 39: 838–843, 2002 [DOI] [PubMed] [Google Scholar]
  • 122.Yoon HE, Ghee JY, Piao S, Song JH, Han DH, Kim S, Ohashi N, Kobori H, Kuro-o M, Yang CW: Angiotensin II blockade upregulates the expression of Klotho, the anti-ageing gene, in an experimental model of chronic cyclosporine nephropathy. Nephrol Dial Transplant 26: 800–813, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lau WL, Leaf EM, Hu MC, Takeno MM, Kuro-o M, Moe OW, Giachelli CM: Vitamin D receptor agonists increase klotho and osteopontin while decreasing aortic calcification in mice with chronic kidney disease fed a high phosphate diet. Kidney Int 82: 1261–1270, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kuczera P, Adamczak M, Wiecek A: Cinacalcet treatment decreases plasma fibroblast growth factor 23 concentration in haemodialysed patients with chronic kidney disease and secondary hyperparathyroidism [published online ahead of print September 20, 2013]. Clin Endocrinol (Oxf) 10.1111/cen.12326 [DOI] [PubMed] [Google Scholar]
  • 125.Kim HJ, Kim H, Shin N, Na KY, Kim YL, Kim D, Chang JH, Song YR, Hwang YH, Kim YS, Ahn C, Lee J, Oh KH, Representing the Cinacalcet stUdy for Peritoneal Dialysis Patients In Double Arm on the Lowing Effect Of iPTH Level (CUPID) Study Group : Cinacalcet lowering of serum fibroblast growth factor-23 concentration may be independent from serum Ca, P, PTH and dose of active vitamin D in peritoneal dialysis patients: A randomized controlled study. BMC Nephrol 14: 112, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Koizumi M, Komaba H, Nakanishi S, Fujimori A, Fukagawa M: Cinacalcet treatment and serum FGF23 levels in haemodialysis patients with secondary hyperparathyroidism. Nephrol Dial Transplant 27: 784–790, 2012 [DOI] [PubMed] [Google Scholar]
  • 127.Wetmore JB, Liu S, Krebill R, Menard R, Quarles LD: Effects of cinacalcet and concurrent low-dose vitamin D on FGF23 levels in ESRD. Clin J Am Soc Nephrol 5: 110–116, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Finch JL, Tokumoto M, Nakamura H, Yao W, Shahnazari M, Lane N, Slatopolsky E: Effect of paricalcitol and cinacalcet on serum phosphate, FGF-23, and bone in rats with chronic kidney disease. Am J Physiol Renal Physiol 298: F1315–F1322, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lavi-Moshayoff V, Wasserman G, Meir T, Silver J, Naveh-Many T: PTH increases FGF23 gene expression and mediates the high-FGF23 levels of experimental kidney failure: A bone parathyroid feedback loop. Am J Physiol Renal Physiol 299: F882–F889, 2010 [DOI] [PubMed] [Google Scholar]
  • 130.Rhee Y, Bivi N, Farrow E, Lezcano V, Plotkin LI, White KE, Bellido T: Parathyroid hormone receptor signaling in osteocytes increases the expression of fibroblast growth factor-23 in vitro and in vivo. Bone 49: 636–643, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Brown WW, Jüppner H, Langman CB, Price H, Farrow EG, White KE, McCormick KL: Hypophosphatemia with elevations in serum fibroblast growth factor 23 in a child with Jansen’s metaphyseal chondrodysplasia. J Clin Endocrinol Metab 94: 17–20, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Saji F, Shigematsu T, Sakaguchi T, Ohya M, Orita H, Maeda Y, Ooura M, Mima T, Negi S: Fibroblast growth factor 23 production in bone is directly regulated by 1{alpha},25-dihydroxyvitamin D, but not PTH. Am J Physiol Renal Physiol 299: F1212–F1217, 2010 [DOI] [PubMed] [Google Scholar]
  • 133.Gutiérrez OM, Smith KT, Barchi-Chung A, Patel NM, Isakova T, Wolf M: (1-34) Parathyroid hormone infusion acutely lowers fibroblast growth factor 23 concentrations in adult volunteers. Clin J Am Soc Nephrol 7: 139–145, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Samadfam R, Richard C, Nguyen-Yamamoto L, Bolivar I, Goltzman D: Bone formation regulates circulating concentrations of fibroblast growth factor 23. Endocrinology 150: 4835–4845, 2009 [DOI] [PubMed] [Google Scholar]
  • 135.López I, Rodríguez-Ortiz ME, Almadén Y, Guerrero F, de Oca AM, Pineda C, Shalhoub V, Rodríguez M, Aguilera-Tejero E: Direct and indirect effects of parathyroid hormone on circulating levels of fibroblast growth factor 23 in vivo. Kidney Int 80: 475–482, 2011 [DOI] [PubMed] [Google Scholar]
  • 136.Burnett-Bowie SM, Henao MP, Dere ME, Lee H, Leder BZ: Effects of hPTH(1–34) infusion on circulating serum phosphate, 1,25-dihydroxyvitamin D, and FGF23 levels in healthy men. J Bone Miner Res 24: 1681–1685, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Hill KM, Martin BR, Wastney ME, McCabe GP, Moe SM, Weaver CM, Peacock M: Oral calcium carbonate affects calcium but not phosphorus balance in stage 3–4 chronic kidney disease. Kidney Int 83: 959–966, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Chertow GM, Block GA, Correa-Rotter R, Drüeke TB, Floege J, Goodman WG, Herzog CA, Kubo Y, London GM, Mahaffey KW, Mix TC, Moe SM, Trotman ML, Wheeler DC, Parfrey PS, EVOLVE Trial Investigators : Effect of cinacalcet on cardiovascular disease in patients undergoing dialysis. N Engl J Med 367: 2482–2494, 2012 [DOI] [PubMed] [Google Scholar]
  • 139.Yanochko GM, Vitsky A, Heyen JR, Hirakawa B, Lam JL, May J, Nichols T, Sace F, Trajkovic D, Blasi E: Pan-FGFR inhibition leads to blockade of FGF23 signaling, soft tissue mineralization, and cardiovascular dysfunction. Toxicol Sci 135: 451–464, 2013 [DOI] [PubMed] [Google Scholar]
  • 140.Heijboer AC, Levitus M, Vervloet MG, Lips P, ter Wee PM, Dijstelbloem HM, Blankenstein MA: Determination of fibroblast growth factor 23. Ann Clin Biochem 46: 338–340, 2009 [DOI] [PubMed] [Google Scholar]
  • 141.Fliser D, Kollerits B, Neyer U, Ankerst DP, Lhotta K, Lingenhel A, Ritz E, Kronenberg F, Kuen E, König P, Kraatz G, Mann JF, Müller GA, Köhler H, Riegler P, MMKD Study Group : Fibroblast growth factor 23 (FGF23) predicts progression of chronic kidney disease: The Mild to Moderate Kidney Disease (MMKD) Study. J Am Soc Nephrol 18: 2600–2608, 2007 [DOI] [PubMed] [Google Scholar]
  • 142.Seiler S, Reichart B, Roth D, Seibert E, Fliser D, Heine GH: FGF-23 and future cardiovascular events in patients with chronic kidney disease before initiation of dialysis treatment. Nephrol Dial Transplant 25: 3983–3989, 2010 [DOI] [PubMed] [Google Scholar]
  • 143.Clinical Laboratory Standards Institute : Defining, Establishing, and Verifying Reference Intervals in the Clinical Laboratory; Approved Guidelines, 3rd Ed., Wayne, PA, Clinical and Laboratory Standards Institute, 2008 [Google Scholar]
  • 144.Seifert ME, de las Fuentes L, Rothstein M, Dietzen DJ, Bierhals AJ, Cheng SC, Ross W, Windus D, Dávila-Román VG, Hruska KA: Effects of phosphate binder therapy on vascular stiffness in early-stage chronic kidney disease. Am J Nephrol 38: 158–167, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Oliveira RB, Cancela AL, Graciolli FG, Dos Reis LM, Draibe SA, Cuppari L, Carvalho AB, Jorgetti V, Canziani ME, Moysés RM: Early control of PTH and FGF23 in normophosphatemic CKD patients: A new target in CKD-MBD therapy? Clin J Am Soc Nephrol 5: 286–291, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Soriano S, Ojeda R, Rodríguez M, Almadén Y, Rodríguez M, Martín-Malo A, Aljama P: The effect of phosphate binders, calcium and lanthanum carbonate on FGF23 levels in chronic kidney disease patients. Clin Nephrol 80: 17–22, 2013 [DOI] [PubMed] [Google Scholar]
  • 147.Koiwa F, Kazama JJ, Tokumoto A, Onoda N, Kato H, Okada T, Nii-Kono T, Fukagawa M, Shigematsu T, ROD21 Clinical Research Group : Sevelamer hydrochloride and calcium bicarbonate reduce serum fibroblast growth factor 23 levels in dialysis patients. Ther Apher Dial 9: 336–339, 2005 [DOI] [PubMed] [Google Scholar]
  • 148.Cancela AL, Oliveira RB, Graciolli FG, dos Reis LM, Barreto F, Barreto DV, Cuppari L, Jorgetti V, Carvalho AB, Canziani ME, Moysés RM: Fibroblast growth factor 23 in hemodialysis patients: Effects of phosphate binder, calcitriol and calcium concentration in the dialysate. Nephron Clin Pract 117: c74–c82, 2011 [DOI] [PubMed] [Google Scholar]
  • 149.Toida T, Fukudome K, Fujimoto S, Yamada K, Sato Y, Chiyotanda S, Kitamura K: Effect of lanthanum carbonate vs. calcium carbonate on serum calcium in hemodialysis patients: A crossover study. Clin Nephrol 78: 216–223, 2012 [DOI] [PubMed] [Google Scholar]
  • 150.Becker GJ, Walker RG, Hewitson TD, Pedagogos E: Phosphate levels—time for a rethink? Nephrol Dial Transplant 24: 2321–2324, 2009 [DOI] [PubMed] [Google Scholar]
  • 151.Ridefelt P, Axelsson J, Larsson A: Diurnal variability of total calcium during normal sleep and after an acute shift of sleep. Clin Chem Lab Med 50: 147–151, 2012 [DOI] [PubMed] [Google Scholar]
  • 152.Portale AA, Halloran BP, Morris RC, Jr: Dietary intake of phosphorus modulates the circadian rhythm in serum concentration of phosphorus. Implications for the renal production of 1,25-dihydroxyvitamin D. J Clin Invest 80: 1147–1154, 1987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Jubiz W, Canterbury JM, Reiss E, Tyler FH: Circadian rhythm in serum parathyroid hormone concentration in human subjects: Correlation with serum calcium, phosphate, albumin, and growth hormone levels. J Clin Invest 51: 2040–2046, 1972 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Jovanovich A, Chonchol M, Cheung AK, Kaufman JS, Greene T, Roberts WL, Smits G, Kendrick J, HOST Investigators : Racial differences in markers of mineral metabolism in advanced chronic kidney disease. Clin J Am Soc Nephrol 7: 640–647, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Gutiérrez OM, Isakova T, Andress DL, Levin A, Wolf M: Prevalence and severity of disordered mineral metabolism in Blacks with chronic kidney disease. Kidney Int 73: 956–962, 2008 [DOI] [PubMed] [Google Scholar]
  • 156.Rejnmark L, Lauridsen AL, Vestergaard P, Heickendorff L, Andreasen F, Mosekilde L: Diurnal rhythm of plasma 1,25-dihydroxyvitamin D and vitamin D-binding protein in postmenopausal women: Relationship to plasma parathyroid hormone and calcium and phosphate metabolism. Eur J Endocrinol 146: 635–642, 2002 [DOI] [PubMed] [Google Scholar]
  • 157.Kao PC, van Heerden JA, Farley DR, Thompson GB, Taylor RL: Intraoperative monitoring of parathyroid hormone with a rapid automated assay that is commercially available. Ann Clin Lab Sci 32: 244–251, 2002 [PubMed] [Google Scholar]
  • 158.el-Hajj Fuleihan G, Klerman EB, Brown EN, Choe Y, Brown EM, Czeisler CA: The parathyroid hormone circadian rhythm is truly endogenous—a general clinical research center study. J Clin Endocrinol Metab 82: 281–286, 1997 [DOI] [PubMed] [Google Scholar]
  • 159.Khosravi A, Cutler CM, Kelly MH, Chang R, Royal RE, Sherry RM, Wodajo FM, Fedarko NS, Collins MT: Determination of the elimination half-life of fibroblast growth factor-23. J Clin Endocrinol Metab 92: 2374–2377, 2007 [DOI] [PubMed] [Google Scholar]
  • 160.Joly D, Drueke TB, Alberti C, Houillier P, Lawson-Body E, Martin KJ, Massart C, Moe SM, Monge M, Souberbielle JC: Variation in serum and plasma PTH levels in second-generation assays in hemodialysis patients: A cross-sectional study. Am J Kidney Dis 51: 987–995, 2008 [DOI] [PubMed] [Google Scholar]
  • 161.English E, McFarlane I, Taylor KP, Halsall DJ: The effect of potassium EDTA on the stability of parathyroid hormone in whole blood. Ann Clin Biochem 44: 297–299, 2007 [DOI] [PubMed] [Google Scholar]
  • 162.Zerwekh JE: The measurement of vitamin D: Analytical aspects. Ann Clin Biochem 41: 272–281, 2004 [DOI] [PubMed] [Google Scholar]
  • 163.Haut LL, Alfrey AC, Guggenheim S, Buddington B, Schrier N: Renal toxicity of phosphate in rats. Kidney Int 17: 722–731, 1980 [DOI] [PubMed] [Google Scholar]
  • 164.Heijboer AC, Blankenstein MA, Kema IP, Buijs MM: Accuracy of 6 routine 25-hydroxyvitamin D assays: Influence of vitamin D binding protein concentration. Clin Chem 58: 543–548, 2012 [DOI] [PubMed] [Google Scholar]
  • 165.Cantor T, Yang Z, Caraiani N, Ilamathi E: Lack of comparability of intact parathyroid hormone measurements among commercial assays for end-stage renal disease patients: Implication for treatment decisions. Clin Chem 52: 1771–1776, 2006 [DOI] [PubMed] [Google Scholar]
  • 166.Almond A, Ellis AR, Walker SW, Scottish Clinical Biochemistry Managed Diagnostic Network : Current parathyroid hormone immunoassays do not adequately meet the needs of patients with chronic kidney disease. Ann Clin Biochem 49: 63–67, 2012 [DOI] [PubMed] [Google Scholar]
  • 167.Souberbielle JC, Roth H, Fouque DP: Parathyroid hormone measurement in CKD. Kidney Int 77: 93–100, 2010 [DOI] [PubMed] [Google Scholar]
  • 168.Brossard JH, Cloutier M, Roy L, Lepage R, Gascon-Barré M, D’Amour P: Accumulation of a non-(1–84) molecular form of parathyroid hormone (PTH) detected by intact PTH assay in renal failure: Importance in the interpretation of PTH values. J Clin Endocrinol Metab 81: 3923–3929, 1996 [DOI] [PubMed] [Google Scholar]
  • 169.Palmer SC, Hayen A, Macaskill P, Pellegrini F, Craig JC, Elder GJ, Strippoli GF: Serum levels of phosphorus, parathyroid hormone, and calcium and risks of death and cardiovascular disease in individuals with chronic kidney disease: A systematic review and meta-analysis. JAMA 305: 1119–1127, 2011 [DOI] [PubMed] [Google Scholar]
  • 170.Block GA, Hulbert-Shearon TE, Levin NW, Port FK: Association of serum phosphorus and calcium x phosphate product with mortality risk in chronic hemodialysis patients: A national study. Am J Kidney Dis 31: 607–617, 1998 [DOI] [PubMed] [Google Scholar]
  • 171.Kestenbaum B, Sampson JN, Rudser KD, Patterson DJ, Seliger SL, Young B, Sherrard DJ, Andress DL: Serum phosphate levels and mortality risk among people with chronic kidney disease. J Am Soc Nephrol 16: 520–528, 2005 [DOI] [PubMed] [Google Scholar]
  • 172.Mehrotra R, Peralta CA, Chen SC, Li S, Sachs M, Shah A, Norris K, Saab G, Whaley-Connell A, Kestenbaum B, McCullough PA, Kidney Early Evaluation Program (KEEP) Investigators : No independent association of serum phosphorus with risk for death or progression to end-stage renal disease in a large screen for chronic kidney disease. Kidney Int 84: 989–997, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Pilz S, Iodice S, Zittermann A, Grant WB, Gandini S: Vitamin D status and mortality risk in CKD: A meta-analysis of prospective studies. Am J Kidney Dis 58: 374–382, 2011 [DOI] [PubMed] [Google Scholar]
  • 174.Shroff R: Phosphate is a vascular toxin. Pediatr Nephrol 28: 583–593, 2013 [DOI] [PubMed] [Google Scholar]
  • 175.Neves KR, Graciolli FG, dos Reis LM, Pasqualucci CA, Moysés RM, Jorgetti V: Adverse effects of hyperphosphatemia on myocardial hypertrophy, renal function, and bone in rats with renal failure. Kidney Int 66: 2237–2244, 2004 [DOI] [PubMed] [Google Scholar]
  • 176.Chonchol M, Kendrick J, Targher G: Extra-skeletal effects of vitamin D deficiency in chronic kidney disease. Ann Med 43: 273–282, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Journal of the American Society of Nephrology : CJASN are provided here courtesy of American Society of Nephrology

RESOURCES