Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Revisiting the Middle Molecule Hypothesis of Uremic Toxicity: A Systematic Review of Beta 2 Microglobulin Population Kinetics and Large Scale Modeling of Hemodialysis Trials In Silico

  • Maria Eleni Roumelioti,

    Affiliation Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, School of Medicine, Albuquerque, NM, United States of America

  • Thomas Nolin,

    Affiliation Department of Pharmacy and Therapeutics, and Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh Schools of Pharmacy and Medicine, Pittsburgh, PA, United States of America

  • Mark L. Unruh,

    Affiliation Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, School of Medicine, Albuquerque, NM, United States of America

  • Christos Argyropoulos

    argchris@hotmail.com

    Affiliation Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, School of Medicine, Albuquerque, NM, United States of America

Abstract

Background

Beta-2 Microglobulin (β2M) is a prototypical “middle molecule” uremic toxin that has been associated with a higher risk of death in hemodialysis patients. A quantitative description of the relative importance of factors determining β2M concentrations among patients with impaired kidney function is currently lacking.

Methods

Herein we undertook a systematic review of existing studies reporting patient level data concerning generation, elimination and distribution of β2M in order to develop a population model of β2M kinetics. We used this model and previously determined relationships between predialysis β2M concentration and survival, to simulate the population distribution of predialysis β2M and the associated relative risk (RR) of death in patients receiving conventional thrice-weekly hemodialysis with low flux (LF) and high flux (HF) dialyzers, short (SD) and long daily (LD) HF hemodialysis sessions and on-line hemodiafiltration at different levels of residual renal function (RRF).

Results

We identified 9 studies of 106 individuals and 156 evaluations of or more compartmental kinetic parameters of β2M. These studies used a variety of experimental methods to determine β2M kinetics ranging from isotopic dilution to profiling of intra/inter dialytic concentration changes. Most of the patients (74/106) were on dialysis with minimal RRF, thus facilitating the estimation of non-renal elimination kinetics of β2M. In large scale (N = 10000) simulations of individuals drawn from the population of β2M kinetic parameters, we found that, higher dialytic removal materially affects β2M exposures only when RRF (renal clearance of β2M) was below 2 ml/min. In patients initiating conventional HF hemodialysis, total loss of RRF was predicted to be associated with a RR of death of more than 20%. Hemodiafiltration and daily dialysis may decrease the high risk of death of anuric patients by 10% relative to conventional, thrice weekly HF dialysis. Only daily long sessions of hemodialysis consistently reduced mortality risk between 7–19% across the range of β2M generation rate.

Conclusions

Preservation of RRF should be considered one of the therapeutic goals of hemodialysis practice. Randomized controlled trials of novel dialysis modalities may require large sample sizes to detect an effect on clinical outcomes even if they enroll anuric patients. The developed population model for β2M may allow personalization of hemodialysis prescription and/or facilitate the design of such studies by identifying patients with higher β2M generation rate.

Introduction

Beta 2 Microglobulin (β2M) is an 11.6 kDa protein expressed in the surface of every nucleated cell, where it non-covalently associates with the alpha-chain of the Major Histocompatibility Complex I (MHC-I)/Human Leukocyte Antigen I (HLA-I) to facilitate antigen presentation. [1,2] It has long been appreciated that glomerular filtration is the major pathway for the elimination of β2M. [37] Residual renal function (RRF), inflammation and malnutrition appear to affect β2M concentration in patients with chronic kidney disease (CKD) [811] and end-stage renal disease (ESRD) [1217]

The main recognized manifestation of β2M accumulation in patients receiving long-term dialysis is dialysis-related amyloidosis [1822], but more recently β2M has also been linked to higher mortality in hemodialysis (HD) patients [14,23,24], to aortic calcification and cardiovascular mortality in patients with non-dialysis dependent CKD [25]. Nevertheless, more efficient dialytic removal of β2M has not equivocally translated into improved outcomes in randomized controlled trials (RCT) of High Flux membranes [14,26] and on-line hemodiafiltration (HDF) [2729] creating uncertainty regarding the clinical effectiveness of enhanced middle molecule removal. Understanding of these discrepant findings and their implication for the middle molecule toxin theory that has been the driving biological hypothesis for the majority of randomized trials to date, requires one to simultaneously consider the ability of the available dialysis modalities to remove β2M and the considerable intra-individual, biological, variability in the kinetics of β2M. However, a quantitative description of the relative importance of intra-individual factors determining β2M concentrations among patients with impaired kidney function is currently lacking.

This report aims to develop a population kinetic model incorporating the intra-individual variability in generation, distribution and extrarenal elimination of β2M, which is then used to describe the disposition of β2M under different HD regimes and levels of RRF. To develop this population kinetic model, we first undertook a patient-level review and synthesis of the literature of clinical studies (either observational or interventional) regarding these kinetic parameters of β2M in humans. We used these parameters to simulate β2M concentrations and relative survival in a population of ESRD patients with different levels of RRF, using previously reported dose response relationships between predialysis β2M concentration and survival [30]. In these simulations we contrasted the intervention protocols utilized in RCTs of HD patients vis-à-vis RRF as determinants of patient survival.

These large scale simulations allowed us to conduct in-silico randomized controlled trials of different dialysis modalities i.e. low flux (LF, negligible β2M clearance) and high flux (HF, higher β2M clearance) membranes in conventional thrice-weekly HD, HF dialysis in short and long daily sessions and HDF. Our simulations not only recapitulate the design and findings of previously reported trials in dialysis, but also generate hypotheses about novel targets of intervention in nephrology and trial designs for the validation of the middle molecule hypothesis.

Materials and Methods

Systematic review of studies of β2M kinetics

These were identified by searching MEDLINE with the following (text) string: “(beta 2 microglobulin) AND (kinetic OR kinetics OR model OR models) AND (mathematical OR compartmental OR compartment OR simulation) AND (volume OR generation OR clearance OR dialysis OR renal OR dialytic OR production)” supplemented by manual inspection of the bibliography of indentified papers in a previous narrative review. [31] We did not screen articles but proceeded to full text of all studies indentified through the Medline search, to exclude those that were review articles, in-vitro or animal investigations, simulation experiments, failure to employ a kinetic model, or reported aggregate rather than subject level data (exclusion criteria). We included papers if they had used a compartmental model to study β2M kinetics, reported patient-level data and were published in English prior to 2015. As this was not an outcomes systematic review, we did not register our systematic review prospectively. Two investigators (MR, and CA) jointly extracted the data (values of kinetic parameters about the generation, intra-compartmental distribution, volume of compartments, non-renal clearance of β2M) from each individual participant in each study using a piloted form. This form and the patient level data extracted from our evidence synthesis are given in S1 Table.

Since classical techniques for the assessment of bias e.g. funnel plots are not applicable when the studies synthesized lacks a discrete health outcome, we were not able to conduct a formal analysis of bias with these methods. However for each study we evaluated the number of parameters reported, those fixed and those unreported by the investigators as an indicator for bias. We considered studies that did not report (or fixed to a specific value) of at most one parameter as studies with minimal risk of bias. As the number of parameters with fixed (or unreported values) increases, the estimated values of the remaining parameters becomes more and more dependent on the specific assumptions of the investigators and thus the risk for bias increases. Further details are provided in the Prisma Checklist (S2 Text).

Compartmental simulation modeling

B2M kinetic simulations were based on the variable volume model [3234] with two compartments (S1 Text and S1 Fig) incorporating inter and intra-dialytic generation, residual renal clearance, non-renal (extrarenal) clearance and dialytic routes of elimination. We simulated the kinetic parameters of 10,000 patients from the scaled–for–weight distributions of the population mean and standard deviation estimated from the literature synthesis at different levels of RRF (0–10 ml/min) and under regimes of conventional, thrice-weekly HD with either LF or HF dialyzers, short and long-daily HF HD and on-line HDF. Dialysis-related parameters reflected the patterns observed in FHN [35,36], HEMO [37] and HDF trials (Dutch CONTRAST [27], Spanish ESHOL [28], Turkish OL-HDF [29]), with specific details in Supplementary Methods in S1 Text. The purpose of these large scale simulations of patients with their unique set of generation/distribution/extrarenal clearance parameters and dialysis settings (dialyzer specifications, treatment time, blood flow rate, ultrafiltration and infusion rate of replacement fluid) was to summarize the effects of different interventions on β2M exposure.

Statistical Analysis

Estimation of population kinetic parameters.

We analyzed studies that collected multiple measurements in the same individual with a bi-level mixed-effect model accounting for individual (first level) and study (second level) heterogeneity; all other studies were analyzed with a random-effects model with a single (study) random effects model. Parameters were log-transformed prior to mixed-effect modeling of the population mean and (log-) variance, which was estimated by the between individual (two level models) or within study (one level model) standard error. Results are reported as means (SE) for the mean and the logarithm of the standard deviation of each log-transformed kinetic parameter. Furthermore, we calculated the population distribution of the untransformed parameters by transforming out of the logarithm and using the properties of the lognormal distribution. The volume of the two distribution compartments of β2M were analyzed both as absolute as scaled (to body weight) numbers. No other analyses e.g. meta-regression were performed on these data.

Analyses of simulation modeling results.

We assessed treatment related exposures to β2M under time-dependent and peak-dependent toxicity perspectives by computing weekly Time Averaged Concentrations (TAC) and mid-weekly pre-dialysis plasma (Cp) concentrations respectively. The relationship between TAC and Cp was analyzed via linear regression for all combinations of dialysis interventions and RRF levels. Dialytic interventions were compared on the basis of both TAC and Cp with a paired t-test. We assumed the following relationship between the relative risk (RR) of death and quintiles of cumulative predialysis β2M concentration observed over seven years in HEMO [30]: 1.0 (β2M< 27.5mg/l, referent), 1.11 (β2M:27.5-35mg/l), 1.35 (β2M:35–42.5mg/l), and 1.50 (β2M>42.5mg/l), obtained by averaging the risk in the last two quintiles due to the plateauing of the risk-exposure curve reported by the original investigators). Intra-individual changes in cumulative predialysis β2M concentration (averaged over the last two weeks of each simulation) between any two regimes in our simulations were thus converted to differences in RR. The latter, averaged over all individuals yield counterfactually the Average Causal Effect (ACE). [38] We employed the connection between the ACE, a statistical measure of cause and effect, and the treatment effect estimated by RCTs [39] to interpret findings of recent trials and suggest hypotheses for testing in future studies. All analyses were performed in R (v2.15.1–3.1.3).

Results

Studies

A PUBMED search identified 57 papers (S2 Table). An additional eight papers were identified by manual searches of the bibliography of the identified papers and a previous narrative review[31]. Four papers out of the fifty identified through database searching fulfilled the criteria for inclusion and met no criteria for exclusion after full text review (see S2 Table for the references of these fifty papers and the inclusion/exclusion status). Three of the eight papers identified through other sources were excluded due to the use of highly compartmentalized, models that could not be reduced to the model utilized in this report [40,41],[42] yielding a total of nine separate studies (Fig 1). Four of the studies were at minimal risk of bias (one or none fixed/unreported parameters). Studies included in the systematic review, used experimental methods ranging from isotopic dilution to profiling of intra/inter dialytic concentration changes (Table 1) to determine β2M kinetics. In total, these studies reported 156 evaluations of one or more parameters of the compartmental model of β2M in 106 patients (Table 1). Most of the patients (74/106) were on dialysis with minimal RRF, facilitating thus the estimation of the non-renal (“extrarenal” [33]) clearance (KER) of β2M. In subjects with normal kidney function, separate estimation of renal clearance (KR) and KER clearances is not possible, so that only a total body clearance (equal to KR+KER) can be estimated.

thumbnail
Fig 1. PRISM flow chart of the systematic review of human studies examining the compartmental kinetics of beta 2 microglobulin.

A total of 58 studies were identified through a database (Medline) and other sources (examination of references of qualifying articles from the literature review, previous narrative review. A total of nine studies met all inclusion and none of the exclusion criteria.

https://doi.org/10.1371/journal.pone.0153157.g001

thumbnail
Table 1. Published patient level data regarding β2 microglobulin kinetics.

https://doi.org/10.1371/journal.pone.0153157.t001

Population β2M Estimates

Parameter estimates, and the resultant population distribution values (median, upper and lower 2.5% tail) are summarized in Table 2. In normal subjects kidney function is the major determinant of total body clearance as the estimated median KR+KER, was equal to 90.43 ml/min vs. 2.92 ml/min for KER in patients on dialysis.

thumbnail
Table 2. β2 microglobulin population kinetic parameters and quantile values.

https://doi.org/10.1371/journal.pone.0153157.t002

Hemodialysis Simulations

Residual renal clearance is a major determinant of β2M concentration in dialysis patients.

The simulated predialysis β2M concentrations of different dialysis regimens at various levels of KR are shown in Fig 2. The highest concentrations were seen in patients on LF HD irrespective of RRF due to minimal dialytic clearance. Conventional HD, short-daily or long-daily HF and HDF resulted in decreased β2M relative to LF HD. Time averaged concentrations (TAC) and predialysis concentrations were highly and positively correlated across all combinations of RRF and dialytic interventions (r2>0.99).

thumbnail
Fig 2. Simulated β2M concentrations at different levels of residual renal clearance and dialysis regimes.

LF: Low Flux Dialysis, HF: High Flux Dialysis, SD: Short Daily Dialysis with High Flux dialyzers (6 times a week, ~ 2 ½ hrs per session), LD: Long Daily Dialysis with High Flux dialyzers (6 times a week, 6 ⅓ hrs per session), HDF = postdilution online hemodiafiltration.

https://doi.org/10.1371/journal.pone.0153157.g002

Regression analysis (Fig 3) also demonstrated a high numerical agreement between the two measures, with the slopes in linear regression being close to unity especially for higher levels of RRF. Nevertheless, this agreement differed for the different modalities; although the TAC was within 8% of the predialysis level for most modality, RRF combinations, it diverged more than 10% in long-daily or HDF at lower RRF levels.

thumbnail
Fig 3. Relation between mid-week simulated predialysis and weekly time averaged β2M concentrations at different levels of residual renal clearance and dialysis regimes.

Correlation coefficients (r2) and regression equations relating the two measures of exposure (gray line) are shown. LF: Low Flux Dialysis, HF: High Flux Dialysis, SD: Short Daily Dialysis with High Flux dialyzers (6 times a week, ~ 2 ½ hrs per session), LD: Long Daily Dialysis with High Flux dialyzers (6 times a week, 6 ⅓ hrs per session), HDF = post dilution online hemodiafiltration. Cp: predialysis plasma concentration, TAC: Time Averaged Concentration.

https://doi.org/10.1371/journal.pone.0153157.g003

Table 3 shows the pair-wise mean differences in β2M concentrations between different regimens at different levels of RRF. It is only in the absence of RRF (KR = 0 ml/min) that higher dialytic removal of β2M in the form of short-daily (SD vs. HF, Table 3), or HDF (HDF vs HF, Table 3) had a substantial effect on β2M relative to conventional HF HD. Similarly, long daily dialysis consistently resulted in lower concentrations of β2M concentrations; the difference between the two modalities widened as renal function declined (LD vs. SD, Table 3). Finally, HDF resulted in predialysis β2M concentration that was very similar to the one achieved with short-daily dialysis at all levels of RRF (HDF vs SD Table 3).

thumbnail
Table 3. Mean pair wise difference in predialysis β2 microglobulin concentration (in mg/L) and associated 95% confidence interval as a function of residual renal clearance.

https://doi.org/10.1371/journal.pone.0153157.t003

Comparisons based on TAC yielded congruent findings with respect to the relative efficiency of the different dialysis regimes and the role of RRF (Table 4). In these comparisons, the absolute difference in TAC (Table 4) between short-daily dialysis and HDF was much larger than the corresponding difference in Cp (shown in Table 3), suggesting that short-daily yields somewhat lower exposures than HDF compared to the expectations based on predialysis concentrations.

thumbnail
Table 4. Mean pair wise difference in the Time Averaged Concentration (TAC) of β2 microglobulin (in mg/L) and associated 95% confidence interval as a function of residual renal clearance.

https://doi.org/10.1371/journal.pone.0153157.t004

Residual renal function is more important than enhanced dialytic removal in determining β2M related outcomes in hemodialysis.

Reduction in residual KR from 10 ml/min to nil resulted in an increase in the percentage of patients whose β2M concentration category (<27.5 mg/l, 27.5–35 mg/l, 35–42.5 mg/l and >42.5 mg/l) increased e.g. 20%-80% in patients receiving HDF and LF respectively. This was associated with an increase in the predicted RR (Fig 4), that was modified by the dialysis regime: 1.32 (LF), 1.21 (HF), 1.13 (SD), 1.03 (LD), and 1.13 (HDF). The magnitude of the RRs far exceeded the reductions in mortality that were predicted on the basis of enhanced dialytic removal of β2M (Fig 5), e.g. less than 8% for comparisons of HF vs. LF, short-daily or long-daily, and HDF vs. LF, HF or short-daily when KR was 2 ml/min. In the absence of kidney function, the smallest RRs were seen in HDF and long-daily vs. LF and HF, and were in the order of 0.85. Under the remaining scenarios, dialysis with a regime that more efficiently removes β2M is predicted to be associated with single digit improvement in the RR.

thumbnail
Fig 4. Simulated changes in β2M and predicted Relative Risk (RR) of death at different levels of renal function.

For each dialysis regime we calculated: i) the percentage of patients undergoing a change in their cumulative predialysis β2M concentration (categorically classified as <27.5 mg/l, 27.5–35 mg/l, 35–42.5 mg/l and >42.5 mg/l) for the different levels of residual renal function (KR) relative to the baseline measurement when KR = 10 ml/min ii) the associated prediction for the RR. Within each dialysis modality, the referent is the state with KR = 10 ml/min

https://doi.org/10.1371/journal.pone.0153157.g004

thumbnail
Fig 5. Simulated changes in β2M and predicted Relative Risk (RR) of death associated with enhanced dialytic removal.

At each level of residual renal function (KR) we calculated: i) the percentage of patients with a change in the cumulative β2M concentration (categorically classified as <27.5 mg/l, 27.5–35 mg/l, 35–42.5 mg/l and >42.5 mg/l) between techniques of higher and lower dialytic removal of β2M ii) the associated prediction for the RR.

https://doi.org/10.1371/journal.pone.0153157.g005

The estimated dialytic effects on β2M category and predicted survival were not uniform across subgroups defined on the basis of quartiles of increasing β2M generation rate. Relative to LF dialysis, adoption of HF membranes would be expected to reduce mortality by more than 10% in patients of the lower two (Q1-2) quartiles (Fig 6), but the effect is smaller and reaches a plateau for higher generation rates. A similar pattern was noted for HDF or short-daily vs. HF dialysis for the Q4 subgroup. Long-daily regimes are anticipated to improve outcomes more for patients at Q2-Q4 (15–19%) rather than those who generate β2M at the lowest generation rate (~7%). As anticipated dialysis effects were quantitatively more important for patients with higher generation at higher levels of RRF (i.e., the RR of 0.95 for short-daily vs. HF was observed for Q1 patients at a KR of 0 ml/min vs. 10 ml/min for Q4 patients).

thumbnail
Fig 6. Simulated changes in β2M and predicted Relative Risk (RR) of death associated with enhanced dialytic removal at different quartiles (Q1-4) of generation rate.

At each level of residual renal function (KR) and quartile of reuse we calculated: i) the percentage of patients with a change in the cumulative β2M concentration (categorically classified as <27.5 mg/l, 27.5–35 mg/l, 35–42.5 mg/l and >42.5 mg/l) between techniques of higher and lower dialytic removal of β2M ii) the associated prediction for the RR.

https://doi.org/10.1371/journal.pone.0153157.g006

Discussion

This report demonstrates estimated predialysis β2M concentrations averaging over the population variability in generation, distribution and extrarenal clearance of β2M under different levels of RRF. Since RRF is a major determinant of β2M in dialysis patients, increased dialytic removal becomes clinically important only when RRF has declined to below 2ml/min. Dialysis effects on survival outcomes related to β2M levels were moderate and heterogeneous in subgroups of patients of different generation rates at all levels of RRF and for a wide spectrum of dialysis regimens.

The population kinetic model reproduces experimental patterns observed in early studies of dialysis patients [13,30,31,5052]. More specifically, we anticipated a large influence of RRF on plasma β2M concentration, particularly when the former declines below 2 ml/min. In retrospect, this prediction is not surprising but we derive this relation from first principles and in a quantitative fashion. This allows our numerical evaluations to generate testable hypotheses that can be verified or refuted by empirical data. In HEMO, predialysis β2M concentrations were higher in anuric patients [30] and declined curvilinearly for higher residual urea clearances (e.g. see Fig 3[30]), a pattern similar to the one described in this report. In the CONTRAST trial of HDF, the average β2M concentration in patients dialyzing with LF membranes and an eGFR of 2.0 ml/min/1.73 m2 at baseline was 30.7–32.3 mg/L, which is in close agreement with our estimate of 29.9 mg/L. [27] At a higher, semi-quantitative level, our analyses suggest that interventions that increase dialytic removal of β2M (i.e., use of HF membranes, addition of convective clearance in the form of HDF, increase in both frequency and duration of treatments) will be masked until RRF is substantially reduced in accordance with reports in HF dialysis [51] and on-line HDF [13,16].

Our findings provide a framework to reconcile reports suggesting that higher dialytic removal (HDF vs. HF) does not have a substantial impact on predialysis β2M concentration [28,29,53]. These discrepancies can be understood by highlighting the importance of both RRF and the (unmeasured) generation rate as determinants of the β2M response to dialysis. These HDF studies enrolled prevalent patients (median time on dialysis: 28–68 months) and with the exception of the study by Ward, [53] provided minimal data on RRF. Hence, in studies inferring the response of β2M to more efficient dialysis one should adjust for these parameters either directly by measuring RRF, or indirectly by using additional patient level covariates that potentially correlate with the generation rate of middle molecules.

Another novel feature of this report is the adoption of a counterfactual perspective in the population-level evaluation of different dialysis regimes. This perspective allowed us to predict relative changes in survival associated with changes in β2M (the prototypical middle molecule [54]) exposure. In ESRD more robust evidence that higher β2M concentrations are associated with worse survival comes from the HEMO study cohort, [23,30] in which β2M levels were assessed prospectively and repeatedly over time. The apparent dose response relationship in HEMO was observed in a cohort of patients with negligible RRF (only 14% of the 1704 patients had RRF >0.75 ml/min at study enrollment), and was detected with time-updated survival models in the presence of extensive multivariable adjustment including RRF. On the other hand, cross-sectional, observational studies utilizing single measurements of β2M yielded partially conflicting associations [24],[55]. More recently, associations have been reported between β2M and survival (all-cause and cardiovascular in NHANES [56] and ARIC [57]), cardiovascular events and calcification in CKD [25], early-onset atherosclerosis in ESRD [58], stroke [59], peripheral arterial disease [6063], and mortality in patients undergoing coronary angiography. [64] Although observational, these associations have held against adjustments for known risk factors and the prevailing level of kidney function (as assessed with cystatin-C or eGFR) suggesting that β2M elevations may have pathologic significance above and beyond its association with glomerular filtration.

These clinical and basic science observations suggest that it is at least possible that β2M may be directly, rather than indirectly, e.g. as a surrogate of RRF, implicated in the heightened morbidity and mortality in ESRD. Thus, combination of the population kinetic model with concentration-effect relationships, known as an exposure-clinical response model in clinical pharmacology [65], can be considered a research tool that facilitates quantitative predictions and testable hypotheses to be generated. One such prediction is that total loss of RRF will be associated with worsened survival in HD. For US patients, who initiate Conventional thrice-weekly HD at an average kidney function of 10 ml/min [66] the RR associated with total loss of kidney function is over 20% (>30% for LF dialysis) in our simulations. This is approximately 60% of the corresponding estimate reported in a prospective Dutch cohort [67]. It is tempting to hypothesize that loss of middle molecule clearance is a large component of the heightened mortality risk due to the loss of RRF observed in the real world, while the remaining excess mortality is explained by the imperfect capability of renal replacement therapies to restore fluid, electrolyte and other uremic toxin (e.g. bound solutes) [6870] homeostasis.

Our simulations predict that higher dialytic removal of β2M will not affect the RR for death until RRF has declined below 2ml/min. This was seen for all comparisons based on interventions that have been tested in actual medium-large scale RCTs: HF vs. LF (HEMO [14] and MPO [26]), SD vs. HF [35], LD vs. HF [36], LF vs. HDF (CONTRAST [27]), HF vs. HDF (ESHOL [28] and Turkish trials [29]) as well as trial configurations that to our knowledge have not been reported in the literature: SD or LD vs. HDF. Furthermore, the benefits at the population level that are likely to accrue due to higher middle molecule removal from such interventions are unlikely to be large relative to the de-facto standard of HF dialysis [71] unless treatment time, frequency and possibly convective clearance [72] are all increased. Overall these predictions are consistent with the findings of RCTs in this area, yet suggest that subgroups defined by toxin generation rate may receive benefit more than others. To our knowledge, this hypothesis has never been evaluated in either a RCT or an associational study context. Hence, the availability of a population kinetic model described herein provides an opportunity to directly test this hypothesis by yielding a tool that can be used to characterize kinetics in an individualized manner and use this information in a research setting. This is similar to the use of quantitative models in pharmacology [73] to estimate patient specific variables that are used to individualize plasma drug concentrations or pharmacodynamic responses in research or clinical care settings. In this light, the observation that the predialysis β2M concentration is numerically very close to the TAC of β2M (at least for dialysis practices that are widely employed in the US), suggests that predialysis β2M concentration monitoring may offer some of the benefits of more extensive modeling.

From a practical standpoint, the numerical simulations reported herein support the argument that delaying loss of kidney function should be counted among the therapeutic HD goals. This argument agrees with a large body of emerging clinical data [74] associating RRF with improved survival, lower hospitalizations, improved anemia and phosphorus management, better volume management and decreased left ventricular hypertrophy. Thus, aiming only for “more dialysis” without considering RRF is too narrow of a focus if better outcomes are to be attained. The recent report that frequent, prolonged HD is associated with faster declines in RRF [75] while short frequent sessions [75] or HDF [76] may have a neutral or even positive impact in sustaining kidney function, suggests that a tailored approach to dialysis prescription that considers both dialytic and kidney function is required.

The findings and interpretations in this report should be viewed in light of certain limitations in the source data, and in the kinetic and outcome models used. Firstly, the available kinetic studies of β2M involved only a small number of patients and it is possible, that previous research failed to include a representative sample from the human population. Furthermore, our search strategy may have failed to identify all relevant publications. Nevertheless, our simulations reproduced a number of experimental findings so that a strong bias from these two sources is unlikely. Secondly, we have assumed that higher dialytic clearance will affect β2M concentrations only through the dialytic removal of the molecule, without affecting its generation rate. Although suggested by some in vitro studies [77,78], other ex vivo [79,80] and in vivo [81] investigations did not demonstrate an effect of dialyzer flux on β2M gene transcription or protein expression, thus providing empiric support for our assumptions regarding this matter. Thirdly, a few studies have raised the possibility that a third compartment [47,82] may be needed to accurately describe β2M kinetic behavior in patients undergoing long-term HD (>6 years). Therefore, in this report we limited our simulations to 3 months, an interval much longer than the 2–3 weeks needed to evaluate the effects of a given dialysis procedure [47], but shorter than the time over which the two-pool model would potentially lose its accuracy. Finally, the association between middle molecules and outcomes was assessed using a single biomarker (β2M).

One possibility is that β2M, although extensively studied, is an imperfect marker for the removal of the entire spectrum of the peptides/proteins of molecular weight >500 kDa classified as middle molecules. [53,72] Notwithstanding the observations that other middle molecules (beta trace protein [83] or cystatin-C [84]) have also been linked to HD outcomes this is a major limitation due to gaps in the existing literature that only further experimental research can resolve. For example, other uremic solute categories (such as the protein bound toxins) may even be more important than middle molecules and these are more efficiently cleared by larger dialyzers in prolonged sessions [68] or by HDF [69,85]. Supporting recent appraisals and criticisms [8688], these considerations suggest the need for a more rigorous evaluation of longitudinal changes in a number of candidate uremic toxins in relation to patient outcomes beyond β2M. Such a task may be facilitated by targeted analyses in existing biorepositories [89,90] established in the context of RCTs or through establishment of collaborative efforts [91] in prospectively followed observational cohorts. [83]

In summary, we have undertaken a quantitative analysis of the available kinetic studies of β2M in order to simulate β2M concentrations and associated changes in survival across a wide range of dialysis regimes ranging from conventional thrice weekly HD to long daily sessions with HF dialyzers and HDF. These simulations support many clinical observations over the last 30 years while suggesting that optimal middle molecule dialysis may critically depend on the preservation of RRF. Future studies should examine the validity of these predictions against non-standard schedules of frequent short and long HD and HDF and test the utility of the estimated population model in individualizing treatment parameters.

Supporting Information

S1 Fig. Compartmental kinetic model of β2M metabolism.

A) Bi-compartmental system describing β2M kinetics consisting of a plasma/perfusing (P) and non-perfusing/non-plasma (NP) with additional material fluxes for patients during hemodialysis sessions (stippled shapes). In each compartment, the symbols V, Φ, C denote the absolute and fractional volume of each compartment and the concentration of β2M respectively. Generation (G) takes place in both compartments, in direct proportion to their fractional volumes. KD, KER, KR are the dialyzer clearance, extrarenal and residual renal clearances. B) System differential equations for patients receiving dialysis (variable volume model). Volume changes during dialysis (Θ = 1) as a result of ultrafiltration (QUF), as well as in the interdialytic intervals (Θ = 0) due to fluid intake (α).

https://doi.org/10.1371/journal.pone.0153157.s001

(TIFF)

S1 Table. Data extraction form and individual subject data extracted from the studies of the systematic review

https://doi.org/10.1371/journal.pone.0153157.s002

(CSV)

S2 Table. Research reports identified through PUBMED search until June 2015.

https://doi.org/10.1371/journal.pone.0153157.s003

(XLS)

S1 Text. Detailed descriptions of the differential equations of the β2M bicompartmental, variable volume model and the numerical aspects of the simulations.

https://doi.org/10.1371/journal.pone.0153157.s004

(DOCX)

S2 Text. PRISMA checklist for the systematic review of studies concerning the kinetics of beta 2 microglobulin.

https://doi.org/10.1371/journal.pone.0153157.s005

(DOC)

Author Contributions

Conceived and designed the experiments: MR TN MLU CA. Performed the experiments: MR. Analyzed the data: CA. Contributed reagents/materials/analysis tools: TN MR. Wrote the paper: MR TN MLU CA.

References

  1. 1. Cox J, Yewdell J, Eisenlohr L, Johnson P, Bennink J. Antigen presentation requires transport of MHC class I molecules from the endoplasmic reticulum. Science. 1990;247: 715–718. pmid:2137259
  2. 2. Perarnau B, Siegrist C-A, Gillet A, Vincent C, Kimura S, Lemonnier FA. [beta]2-Microglobulin restriction of antigen presentation. Nature. 1990;346: 751–754. pmid:1697039
  3. 3. Ravnskov U, Johansson BG, Göthlin J. Renal extraction of 2 -microglobulin. Scand J Clin Lab Invest. 1972;30: 71–75. pmid:4116283
  4. 4. Wibell LB. Studies on beta2-microglobulin in patients and normal subjects. Acta Clin Belg. 1976;31: 14–26. pmid:65885
  5. 5. Bailey RR, Tisch GW, Pearson S. Serum beta2-microglobulin in the assessment of renal function. N Z Med J. 1978;87: 168–170. pmid:77514
  6. 6. Karlsson FA, Wibell L, Evrin PE. beta 2-Microglobulin in clinical medicine. Scand J Clin Lab Invest Suppl. 1980;154: 27–37. pmid:6163193
  7. 7. François B, Assenat H, Cahen R, Bizollon C. [Is the serum level of beta 2 microglobulin a better marker of glomerular filtration than blood creatinine?]. Nephrologie. 1982;3: 116–118. pmid:6183598
  8. 8. Jung K, Schulze BD, Sydow K, Pergande M, Precht K, Schreiber G. Diagnostic value of low-molecular mass proteins in serum for the detection of reduced glomerular filtration rate. J Clin Chem Clin Biochem. 1987;25: 499–503. pmid:3320263
  9. 9. Acchiardo S, Kraus AP Jr, Jennings BR. Beta 2-microglobulin levels in patients with renal insufficiency. Am J Kidney Dis. 1989;13: 70–74. pmid:2643313
  10. 10. Donadio C, Lucchesi A, Ardini M, Giordani R. Cystatin C, beta 2-microglobulin, and retinol-binding protein as indicators of glomerular filtration rate: comparison with plasma creatinine. J Pharm Biomed Anal. 2001;24: 835–842. pmid:11248475
  11. 11. Bianchi C, Donadio C, Tramonti G, Consani C, Lorusso P, Rossi G. Reappraisal of serum beta2-microglobulin as marker of GFR. Ren Fail. 2001;23: 419–429. pmid:11499557
  12. 12. Brown PH, Kalra PA, Turney JH, Cooper EH. Serum low-molecular-weight proteins in haemodialysis patients: effect of residual renal function. Nephrol Dial Transplant. 1988;3: 169–173. pmid:2459634
  13. 13. Fry AC, Singh DK, Chandna SM, Farrington K. Relative Importance of Residual Renal Function and Convection in Determining Beta-2-Microglobulin Levels in High-Flux Haemodialysis and On-Line Haemodiafiltration. Blood Purif. 2007;25: 295–302. pmid:17622712
  14. 14. Eknoyan G, Beck GJ, Cheung AK, Daugirdas JT, Greene T, Kusek JW, et al. Effect of Dialysis Dose and Membrane Flux in Maintenance Hemodialysis. N Engl J Med. 2002;347: 2010–2019. pmid:12490682
  15. 15. Amici G, Virga G, Da Rin G, Grandesso S, Vianello A, Gatti P, et al. Serum beta-2-microglobulin level and residual renal function in peritoneal dialysis. Nephron. 1993;65: 469–471. pmid:8290003
  16. 16. Penne EL, van der Weerd NC, Blankestijn PJ, van den Dorpel MA, Grooteman MPC, Nubé MJ, et al. Role of residual kidney function and convective volume on change in beta2-microglobulin levels in hemodiafiltration patients. Clin J Am Soc Nephrol. 2010;5: 80–86. pmid:19965537
  17. 17. Cianciolo G, Colí L, La Manna G, Donati G, D’Addio F, Comai G, et al. Is beta2-microglobulin-related amyloidosis of hemodialysis patients a multifactorial disease? A new pathogenetic approach. Int J Artif Organs. 2007;30: 864–878. pmid:17992647
  18. 18. Jadoul M, Garbar C, van Ypersele de Strihou C. Pathological aspects of beta(2)-microglobulin amyloidosis. Semin Dial. 2001;14: 86–89. pmid:11264772
  19. 19. Kiss E, Keusch G, Zanetti M, Jung T, Schwarz A, Schocke M, et al. Dialysis-related amyloidosis revisited. AJR Am J Roentgenol. 2005;185: 1460–1467. pmid:16303998
  20. 20. Danesh F, Ho LT. Dialysis-related amyloidosis: history and clinical manifestations. Semin Dial. 2001;14: 80–85. pmid:11264771
  21. 21. Yamamoto S, Gejyo F. Historical background and clinical treatment of dialysis-related amyloidosis. Biochim Biophys Acta. 2005;1753: 4–10. pmid:16226497
  22. 22. Kazama JJ, Yamamoto S, Takahashi N, Ito Y, Maruyama H, Narita I, et al. Abeta-2M-amyloidosis and related bone diseases. J Bone Miner Metab. 2006;24: 182–184. pmid:16502130
  23. 23. Cheung AK, Greene T, Leypoldt JK, Yan G, Allon M, Delmez J, et al. Association between Serum β2-Microglobulin Level and Infectious Mortality in Hemodialysis Patients. Clin J Am Soc Nephrol. 2008;3: 69–77. pmid:18057309
  24. 24. Okuno S, Ishimura E, Kohno K, Fujino-Katoh Y, Maeno Y, Yamakawa T, et al. Serum beta2-microglobulin level is a significant predictor of mortality in maintenance haemodialysis patients. Nephrol Dial Transplant. 2009;24: 571–577. pmid:18799606
  25. 25. Liabeuf S, Lenglet A, Desjardins L, Neirynck N, Glorieux G, Lemke HD, et al. Plasma beta-2 microglobulin is associated with cardiovascular disease in uremic patients. Kidney Int. 2012;82: 1297–303. pmid:22895515
  26. 26. Locatelli F, Martin-Malo A, Hannedouche T, Loureiro A, Papadimitriou M, Wizemann V, et al. Effect of membrane permeability on survival of hemodialysis patients. J Am Soc Nephrol. 2009;20: 645–54. pmid:19092122
  27. 27. Grooteman MPC, van den Dorpel MA, Bots ML, Penne EL, van der Weerd NC, Mazairac AHA, et al. Effect of online hemodiafiltration on all-cause mortality and cardiovascular outcomes. J Am Soc Nephrol. 2012;23: 1087–1096. pmid:22539829
  28. 28. Maduell F, Moreso F, Pons M, Ramos R, Mora-Macia J, Carreras J, et al. High-efficiency postdilution online hemodiafiltration reduces all-cause mortality in hemodialysis patients. J Am Soc Nephrol. 2013;24: 487–97. pmid:23411788
  29. 29. Ok E, Asci G, Toz H, Ok ES, Kircelli F, Yilmaz M, et al. Mortality and cardiovascular events in online haemodiafiltration (OL-HDF) compared with high-flux dialysis: results from the Turkish OL-HDF Study. Nephrol Dial Transplant. 2013;28: 192–202. pmid:23229932
  30. 30. Cheung AK, Rocco MV, Yan G, Leypoldt JK, Levin NW, Greene T, et al. Serum beta-2 Microglobulin Levels Predict Mortality in Dialysis Patients: Results of the HEMO Study. J Am Soc Nephrol. 2006;17: 546–555. pmid:16382021
  31. 31. Winchester JF, Salsberg JA, Levin NW. Beta-2 microglobulin in ESRD: an in-depth review. Advances in Renal Replacement Therapy. 2003;10: 279–309. pmid:14681859
  32. 32. Stiller S, Xu XQ, Gruner N, Vienken J, Mann H. Validation of a two-pool model for the kinetics of beta2-microglobulin. Int J Artif Organs. 2002;25: 411–420. pmid:12074339
  33. 33. Ward RA, Greene T, Hartmann B, Samtleben W. Resistance to intercompartmental mass transfer limits beta2-microglobulin removal by post-dilution hemodiafiltration. Kidney Int. 2006;69: 1431–1437. pmid:16395268
  34. 34. Kanamori T, Sakai K. An estimate of beta 2-microglobulin deposition rate in uremic patients on hemodialysis using a mathematical kinetic model. Kidney Int. 1995;47: 1453–1457. pmid:7637274
  35. 35. Chertow GM, Levin NW, Beck GJ, Depner TA, Eggers PW, Gassman JJ, et al. In-center hemodialysis six times per week versus three times per week. N Engl J Med. 2010;363: 2287–2300. pmid:21091062
  36. 36. Rocco MV, Lockridge RS Jr, Beck GJ, Eggers PW, Gassman JJ, Greene T, et al. The effects of frequent nocturnal home hemodialysis: the Frequent Hemodialysis Network Nocturnal Trial. Kidney Int. 2011;80: 1080–1091. pmid:21775973
  37. 37. CHEUNG AK, AGODOA LY, DAUGIRDAS JT, DEPNER TA, GOTCH FA, GREENE T, et al. Effects of Hemodialyzer Reuse on Clearances of Urea and {beta}2-Microglobulin. J Am Soc Nephrol. 1999;10: 117–127. pmid:9890317
  38. 38. Rubin DB. Estimating causal effects of treatments in randomized and nonrandomized studies. Journal of Educational Psychology. 1974;66: 688–701.
  39. 39. Pearl J. Causal inference in statistics: An overview. Statist Surv. 2009;3: 96–146.
  40. 40. Gotch F, Levin N, Zasuwa G, Tayeb J. Kinetics of beta-2-microglobulin in hemodialysis. Contrib Nephrol. 1989;74: 132–138. pmid:2702130
  41. 41. Lee CJ, Hsiong CH, Chang YL, Cheng CH, Lian JD. Statistical and parametric analysis of beta-2-microglobulin removal from uremic patients in high flux hemodialysis. ASAIO J. 1994;40: 62–66. pmid:8186494
  42. 42. Takesawa S, Hidai S, Ogawa H, Takagi T, Saito A, Sakai K. Kinetic analysis of beta-2-microglobulin behavior for hemodialysis patients. Jpn J Artif Organs. 1988;17: 42–45.
  43. 43. Karlsson FA, Groth T, Sege K, Wibell L, Peterson PA. Turnover in humans of beta 2-microglobulin: the constant chain of HLA-antigens. Eur J Clin Invest. 1980;10: 293–300. pmid:6159213
  44. 44. Vincent C, Pozet N, Revillard JP. Plasma beta 2 microglobulin turnover in renal insufficiency. Acta Clin Belg Suppl. 1980;35: 2–12.
  45. 45. Maeda K, Shinzato T, Ota T, Kobayakawa H, Takai I, Fujita Y, et al. Beta-2-microglobulin generation rate and clearance rate in maintenance hemodialysis patients. Nephron. 1990;56: 118–125. pmid:2243566
  46. 46. Floege J, Bartsch A, Schulze M, Shaldon S, Koch KM, Smeby LC. Clearance and synthesis rates of beta 2-microglobulin in patients undergoing hemodialysis and in normal subjects. J Lab Clin Med. 1991;118: 153–165. pmid:1856578
  47. 47. Odell RA, Slowiaczek P, Moran JE, Schindhelm K. Beta 2-microglobulin kinetics in end-stage renal failure. Kidney Int. 1991;39: 909–919. pmid:2067207
  48. 48. Vincent C, Chanard J, Caudwell V, Lavaud S, Wong T, Revillard JP. Kinetics of 125I-beta 2-microglobulin turnover in dialyzed patients. Kidney Int. 1992;42: 1434–1443. pmid:1474777
  49. 49. Xu XQ, Gruner N, Al-Bashir A, Trutt-Ibing CH, Melzer H, Fassbinder W, et al. Determination of extra renal clearance and generation rate of beta2-microglobulin in hemodialysis patients using a kinetic model. ASAIO J. 2001;47: 623–627. pmid:11730200
  50. 50. Wizemann V, Lotz C, Techert F, Uthoff S. On-line haemodiafiltration versus low-flux haemodialysis. A prospective randomized study. Nephrol Dial Transplant. 2000;15 Suppl 1: 43–48. pmid:10737166
  51. 51. McCarthy JT, Williams AW, Johnson WJ. Serum beta 2-microglobulin concentration in dialysis patients: importance of intrinsic renal function. J Lab Clin Med. 1994;123: 495–505. pmid:8144998
  52. 52. Leypoldt JK, Cheung AK, Deeter RB. Single compartment models for evaluating beta 2-microglobulin clearance during hemodialysis. ASAIO J. 1997;43: 904–909. pmid:9386841
  53. 53. Ward RA, Schmidt B, Hullin J, Hillebrand GF, Samtleben W. A Comparison of On-Line Hemodiafiltration and High-Flux Hemodialysis: A Prospective Clinical Study. JASN. 2000;11: 2344–2350. pmid:11095657
  54. 54. Tattersall J. Clearance of beta-2-microglobulin and middle molecules in haemodiafiltration. Contributions to nephrology. 2007;158: 201–9. pmid:17684359
  55. 55. Kim KM, Kim S-S, Kim H, Koo T, Im EY, Kim SB. Higher serum beta2-microglobulin levels are associated with better survival in chronic hemodialysis patients: a reverse epidemiology. Clin Nephrol. 2011;75: 458–465. pmid:21543026
  56. 56. Foster MC, Inker LA, Levey AS, Selvin E, Eckfeldt J, Juraschek SP, et al. Novel Filtration Markers as Predictors of All-Cause and Cardiovascular Mortality in US Adults. Am J Kidney Dis. 2013; Available: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=23518194
  57. 57. Astor BC, Shafi T, Hoogeveen RC, Matsushita K, Ballantyne CM, Inker LA, et al. Novel markers of kidney function as predictors of ESRD, cardiovascular disease, and mortality in the general population. Am J Kidney Dis. 2012;59: 653–662. pmid:22305758
  58. 58. Zumrutdal A, Sezer S, Demircan S, Seydaoglu G, Ozdemir FN, Haberal M. Cardiac troponin I and beta 2 microglobulin as risk factors for early-onset atherosclerosis in patients on haemodialysis. Nephrology (Carlton). 2005;10: 453–458.
  59. 59. Amighi J, Hoke M, Mlekusch W, Schlager O, Exner M, Haumer M, et al. Beta 2 microglobulin and the risk for cardiovascular events in patients with asymptomatic carotid atherosclerosis. Stroke. 2011;42: 1826–33. pmid:21546482
  60. 60. Wilson AM, Kimura E, Harada RK, Nair N, Narasimhan B, Meng XY, et al. Beta2-microglobulin as a biomarker in peripheral arterial disease: proteomic profiling and clinical studies. Circulation. 2007;116: 1396–403. pmid:17724262
  61. 61. Fung ET, Wilson AM, Zhang F, Harris N, Edwards KA, Olin JW, et al. A biomarker panel for peripheral arterial disease. Vasc Med. 2008;13: 217–224. pmid:18687758
  62. 62. Kals J, Zagura M, Serg M, Kampus P, Zilmer K, Unt E, et al. β2-microglobulin, a novel biomarker of peripheral arterial disease, independently predicts aortic stiffness in these patients. Scand J Clin Lab Invest. 2011;71: 257–263. pmid:21314441
  63. 63. Hiatt WR, Zakharyan A, Fung ET, Crutcher G, Smith A, Stanford C, et al. A validated biomarker panel to identify peripheral artery disease. Vasc Med. 2012;17: 386–393. pmid:23086582
  64. 64. Nead KT, Zhou MJ, Caceres RD, Sharp SJ, Wehner MR, Olin JW, et al. Usefulness of the addition of beta-2-microglobulin, cystatin C and C-reactive protein to an established risk factors model to improve mortality risk prediction in patients undergoing coronary angiography. Am J Cardiol. 2013;111: 851–856. pmid:23290308
  65. 65. Zhang L, Pfister M, Meibohm B. Concepts and Challenges in Quantitative Pharmacology and Model-Based Drug Development. AAPS J. 2008;10: 552–559. pmid:19003542
  66. 66. USRDS 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.
  67. 67. van der Wal WM, Noordzij M, Dekker FW, Boeschoten EW, Krediet RT, Korevaar JC, et al. Full loss of residual renal function causes higher mortality in dialysis patients; findings from a marginal structural model. Nephrol Dial Transplant. 2011;26: 2978–2983. pmid:21317411
  68. 68. Sirich TL, Luo FJ-G, Plummer NS, Hostetter TH, Meyer TW. Selectively increasing the clearance of protein-bound uremic solutes. Nephrol Dial Transplant. 2012;27: 1574–1579. pmid:22231033
  69. 69. Eloot S, Dhondt A, Van Landschoot M, Waterloos M- A, Vanholder R. Removal of water-soluble and protein-bound solutes with reversed mid-dilution versus post-dilution haemodiafiltration. Nephrol Dial Transplant. 2012;27: 3278–3283. pmid:22492823
  70. 70. Bammens B, Evenepoel P, Verbeke K, Vanrenterghem Y. Removal of middle molecules and protein-bound solutes by peritoneal dialysis and relation with uremic symptoms. Kidney Int. 2003;64: 2238–2243. pmid:14633148
  71. 71. Blankestijn PJ, Ledebo I, Canaud B. Hemodiafiltration: clinical evidence and remaining questions. Kidney Int. 2010;77: 581–7. pmid:20130529
  72. 72. Maduell F, Arias M, Durán CE, Vera M, Fontseré N, Azqueta M, et al. Nocturnal, every-other-day, online haemodiafiltration: an effective therapeutic alternative. Nephrol Dial Transplant. 2012;27: 1619–1631. pmid:21931125
  73. 73. Lesko LJ, Schmidt S. Individualization of drug therapy: history, present state, and opportunities for the future. Clin Pharmacol Ther. 2012;92: 458–466. pmid:22948891
  74. 74. Vilar E, Farrington K. Emerging importance of residual renal function in end-stage renal failure. Semin Dial. 2011;24: 487–494. pmid:21999737
  75. 75. Daugirdas JT, Greene T, Rocco MV, Kaysen GA, Depner TA, Levin NW, et al. Effect of frequent hemodialysis on residual kidney function. Kidney Int. 2013
  76. 76. Schiffl H, Lang SM, Fischer R. Effects of high efficiency post-dilution on-line hemodiafiltration or conventional hemodialysis on residual renal function and left ventricular hypertrophy. Int Urol Nephrol. 2012;
  77. 77. Jahn B, Betz M, Deppisch R, Janssen O, Hänsch GM, Ritz E. Stimulation of beta 2-microglobulin synthesis in lymphocytes after exposure to Cuprophan dialyzer membranes. Kidney Int. 1991;40: 285–290. pmid:1942777
  78. 78. Schoels M, Jahn B, Hug F, Deppisch R, Ritz E, Hänsch GM. Stimulation of mononuclear cells by contact with cuprophan membranes: further increase of beta 2-microglobulin synthesis by activated late complement components. Am J Kidney Dis. 1993;21: 394–399. pmid:8465819
  79. 79. Campistol JM, Molina R, Bernard DB, Rodriguez R, Mirapeix E, Munoz-Gomez JM, et al. Synthesis of beta 2-microglobulin in lymphocyte culture: role of hemodialysis, dialysis membranes, dialysis-amyloidosis, and lymphokines. Am J Kidney Dis. 1993;22: 691–699. pmid:8238015
  80. 80. Zingraff J, Beyne P, Ureña P, Uzan M, Man Nguyen Khoa, Descamps-Latscha B, et al. Influence of haemodialysis membranes on beta 2-microglobulin kinetics: in vivo and in vitro studies. Nephrol Dial Transplant. 1988;3: 284–290. pmid:3140102
  81. 81. Haufe CC, Eismann U, Deppisch RM, Stein G. Expression of beta2-microglobulin and c-fos mRNA: is there an influence of high- or low-flux dialyzer membranes? Kidney Int Suppl. 2001;78: S177–181. pmid:11169006
  82. 82. Chanard J, Vincent C, Caudwell V, Lavaud S, Toupance O, Wong T, et al. Beta 2-microglobulin metabolism in uremic patients who are undergoing dialysis. Kidney Int Suppl. 1993;41: S83–87. pmid:8320953
  83. 83. Shafi T, Parekh RS, Jaar BG, Plantinga LC, Oberai PC, Eckfeldt JH, et al. Serum β-trace protein and risk of mortality in incident hemodialysis patients. Clin J Am Soc Nephrol. 2012;7: 1435–1445. pmid:22745274
  84. 84. Shin MJ, Song SH, Kwak IS, Lee SB, Lee DW, Seong EY, et al. Serum cystatin C as a predictor for cardiovascular events in end-stage renal disease patients at the initiation of dialysis. Clin Exp Nephrol. 2012;16: 456–463. pmid:22278599
  85. 85. Meert N, Eloot S, Schepers E, Lemke H-D, Dhondt A, Glorieux G, et al. Comparison of removal capacity of two consecutive generations of high-flux dialysers during different treatment modalities. Nephrol Dial Transplant. 2011;26: 2624–2630. pmid:21310741
  86. 86. Vanholder R, Van Laecke S, Glorieux G. The middle-molecule hypothesis 30 years after: lost and rediscovered in the universe of uremic toxicity? J Nephrol. 2008;21: 146–160. pmid:18446708
  87. 87. Eloot S, Van Biesen W, Vanholder R. A sad but forgotten truth: the story of slow-moving solutes in fast hemodialysis. Semin Dial. 2012;25: 505–9. pmid:22925227
  88. 88. Dobre M, Meyer TW, Hostetter TH. Searching for uremic toxins. Clin J Am Soc Nephrol. 2013;8: 322–327. pmid:23024165
  89. 89. Pan H, Ardini MA, Bakalov V, Delatte M, Eggers P, Ganapathi L, et al. “What”s in the NIDDK CDR?’—public query tools for the NIDDK central data repository. Database (Oxford). 2013; bas058.
  90. 90. Turner CF, Pan H, Silk GW, Ardini MA, Bakalov V, Bryant S, et al. The NIDDK Central Repository at 8 years—ambition, revision, use and impact. Database (Oxford). 2011; bar043.
  91. 91. Vanholder R, Abou-Deif O, Argiles A, Baurmeister U, Beige J, Brouckaert P, et al. The role of EUTox in uremic toxin research. Semin Dial. 2009;22: 323–328. pmid:19708975