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

Systematic review of international studies evaluating MDRD and CKD-EPI estimated glomerular filtration rate (eGFR) equations in Black adults

  • Ebele M. Umeukeje ,

    Contributed equally to this work with: Ebele M. Umeukeje, Taneya Y. Koonce

    Roles Conceptualization, Supervision, Writing – original draft, Writing – review & editing

    Affiliations Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America, Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • Taneya Y. Koonce ,

    Contributed equally to this work with: Ebele M. Umeukeje, Taneya Y. Koonce

    Roles Conceptualization, Investigation, Project administration, Supervision, Visualization, Writing – original draft, Writing – review & editing

    taneya.koonce@vumc.org

    Affiliation Center for Knowledge Management, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • Sheila V. Kusnoor,

    Roles Conceptualization, Investigation, Project administration, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Center for Knowledge Management, Vanderbilt University Medical Center, Nashville, TN, United States of America, Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • Ifeoma I. Ulasi,

    Roles Conceptualization, Supervision, Writing – review & editing

    Affiliation Renal Unit, Department of Medicine, College of Medicine, University of Nigeria/University of Nigeria Teaching Hospital, Ituku-Ozalla, Nigeria

  • Sophia Kostelanetz,

    Roles Conceptualization, Supervision, Writing – original draft, Writing – review & editing

    Affiliation Division of General Internal Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • Annette M. Williams,

    Roles Conceptualization, Investigation, Project administration, Visualization, Writing – review & editing

    Affiliation Center for Knowledge Management, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • Mallory N. Blasingame,

    Roles Conceptualization, Investigation, Project administration, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Center for Knowledge Management, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • Marcia I. Epelbaum,

    Roles Conceptualization, Investigation, Writing – review & editing

    Affiliation Center for Knowledge Management, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • Dario A. Giuse,

    Roles Conceptualization, Investigation, Writing – review & editing

    Affiliation Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • Annie N. Apple,

    Roles Conceptualization, Writing – review & editing

    Affiliation Vanderbilt University School of Medicine, Nashville, TN, United States of America

  • Karampreet Kaur,

    Roles Conceptualization, Writing – review & editing

    Affiliation Vanderbilt University School of Medicine, Nashville, TN, United States of America

  • Tavia González Peña,

    Roles Conceptualization, Writing – review & editing

    Affiliation Vanderbilt University School of Medicine, Nashville, TN, United States of America

  • Danika Barry,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Obstetrics & Gynecology, McGaw Medical Center of Northwestern University, Chicago, IL, United States of America

  • Leo G. Eisenstein,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Medicine, NYU Langone Medical Center, New York, NY, United States of America

  • Cameron T. Nutt,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States of America

  •  [ ... ],
  • Nunzia B. Giuse

    Roles Conceptualization, Investigation, Project administration, Supervision, Writing – original draft, Writing – review & editing

    Affiliations Center for Knowledge Management, Vanderbilt University Medical Center, Nashville, TN, United States of America, Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States of America, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • [ view all ]
  • [ view less ]

Abstract

Use of race adjustment in estimating glomerular filtration rate (eGFR) has been challenged given concerns that it may negatively impact the clinical care of Black patients, as it results in Black patients being systematically assigned higher eGFR values than non-Black patients. We conducted a systematic review to assess how well eGFR, with and without race adjustment, estimates measured GFR (mGFR) in Black adults globally. A search across multiple databases for articles published from 1999 to May 2021 that compared eGFR to mGFR and reported outcomes by Black race was performed. We included studies that assessed eGFR using the Modification of Diet in Renal Disease (MDRD) and Chronic Kidney Disease Epidemiology Collaboration (CKD-EPICr) creatinine equations. Risk of study bias and applicability were assessed with the QUality Assessment of Diagnostic Accuracy Studies-2. Of 13,167 citations identified, 12 met the data synthesis criteria (unique patient cohorts in which eGFR was compared to mGFR with and without race adjustment). The studies included patients with and without kidney disease from Africa (n = 6), the United States (n = 3), Europe (n = 2), and Brazil (n = 1). Of 11 CKD-EPI equation studies, all assessed bias, 8 assessed accuracy, 6 assessed precision, and 5 assessed correlation/concordance. Of 7 MDRD equation studies, all assessed bias, 6 assessed accuracy, 5 assessed precision, and 3 assessed correlation/concordance. The majority of studies found that removal of race adjustment improved bias, accuracy, and precision of eGFR equations for Black adults. Risk of study bias was often unclear, but applicability concerns were low. Our systematic review supports the need for future studies to be conducted in diverse populations to assess the possibility of alternative approaches for estimating GFR. This study additionally provides systematic-level evidence for the American Society of Nephrology—National Kidney Foundation Task Force efforts to pursue other options for GFR estimation.

Introduction

Accurate assessment of kidney function is essential for proper diagnosis, staging, and management of chronic kidney disease (CKD). The gold standard for evaluating kidney function is measured glomerular filtration rate (mGFR), which relies on infusing chemicals, such as iothalamate, into the blood and quantifying urine clearance. Estimated GFR (eGFR) is based on the measurement of serum filtration markers, such as creatinine or cystatin C, and therefore is more practical to obtain in clinical practice [1].

The Modification of Diet in Renal Disease (MDRD) and Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equations are commonly used in the United States (U.S.) and internationally. Each includes serum creatinine measurement as a key variable for determining eGFR [24]. The equations were developed in large U.S. cohorts and include race, sex, and age variables [5]. Use of the race variable increases reported eGFR by 21.2% (MDRD) or 15.9% (CKD-EPI) in Black patients [3, 6].

While sex and age are biological variables, the meaning and classification of race have evolved over time. Definitions of race are often inconsistently applied, and the utility of race adjustment in non-U.S. populations is unclear [7]. The appropriateness of including race in clinical algorithms has been questioned given that race is a sociopolitical rather than biological construct and the urgent need for clinical medicine to confront structural racism in our practices [714].

In the U.S., the prevalence of chronic kidney disease (CKD) and end-stage renal disease is higher among Black Americans relative to White Americans [15, 16]. While genetic and social factors play a large role in kidney health disparities in the U.S., inappropriately including race in eGFR equations can increase health inequities [14, 17]. Race-adjusted glomerular filtration rate (GFR) estimates could delay clinical care dependent on eGFR thresholds, such as specialist referrals or transplantation, and impact dosing decisions [8, 9, 14].

Concerns about health equity and the lack of evidence for race adjustment led several institutions to omit race from eGFR equations [18]. Some researchers cautioned about unintended consequences, including the potential for Black patients to be overdiagnosed and overtreated, or prematurely deprescribed medications such as metformin or empagliflozin [8, 11, 14, 18]. In July 2020, the National Kidney Foundation and the American Society of Nephrology (NKF-ASN) created a task force to evaluate the inclusion of race in eGFR equations in response to growing controversy [19]. The task force ultimately recommended a new 2021 CKD-EPI creatinine equation refit without the race variable for U.S. adults [20, 21].

A systematic review of the global literature on the use of eGFR equations for Black patients is needed to inform clinical decision-making worldwide [14] and strengthen the adoption of the NKF-ASN task force recommendations. Our team conducted a systematic review to provide the highest level of evidence for assessing the use of eGFR race adjustment in Black patients. We aimed to answer the question: How well does eGFR, with and without race adjustment, predict mGFR in Black adults in the U.S. and internationally?

Methods

Protocol

A protocol with investigation parameters, data extraction procedures, and quality assessment processes was used to guide the review process (S1 Table). The protocol and systematic review are reported following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines (S2 Table) [2224].

Eligibility criteria

Inclusion criteria were validation and/or comparison studies that included Black adults (ages 18 and older), compared an eGFR formula to mGFR or to another eGFR formula, and were published in English, French, Spanish, or Portuguese. All research article types were eligible, including conference abstracts. Items were excluded if they did not use mGFR as a reference test as assessed by 51Cr-EDTA, inulin, 125I-iothalamate, iohexol, or 99mTc-DTPA exogenous markers, did not use either an MDRD or CKD-EPI eGFR formula as an index test, did not use isotope dilution mass spectrometry (IDMS) traceable creatinine assays, or did not report outcomes by Black race. Conference abstracts published prior to 2015 were excluded as the findings would likely have been later published as journal articles. Studies reporting preliminary results were excluded when a more complete publication with the final results was identified.

Search process

PubMed, Embase (via OVID), ScienceDirect (Elsevier), Web of Science, and Google Scholar (via the Publish or Perish software [25]) were searched for citations published after January 1, 1999. Database searches, originally conducted on October 23, 2020, were re-executed on May 6, 2021. All search results were deduplicated based on unique identifiers (e.g., digital object identifier, article title). See S1 File for the full search strategies and search process details.

Study screening and data extraction

The abstracts and full-text of each publication were independently screened by 2 reviewers. Decisions were collected via REDCap® [26] forms created by the study team. Discordant ratings between reviewers were resolved using an independent third person. Data extraction was also conducted independently by 2 reviewers for each included study meeting eligibility criteria; a third person adjudicated non-consensus ratings. Data extracted included: citation details, country of study, study aims, total participants, population characteristics (e.g., demographics, kidney disease status), study eligibility criteria, eGFR and mGFR calculation methods, type of eGFR equation used, eGFR as assessed with and/or without race coefficient in Black adults, mGFR in Black adults, performance measure results, and statistical significance.

The main outcomes included bias (defined as the difference between eGFR and mGFR), accuracy (proportion of patients with eGFR values within a given threshold, such as 30% of mGFR, also referred to as P30), precision, and correlation/concordance of eGFR and mGFR. Additional outcomes were reporting of social determinants of health (e.g., income, educational level, lifestyle choices) and non-GFR determinants of creatinine: underlying co-morbidities (e.g., liver disease, anorexia, chronic illnesses), and medication use that can lead to false elevation of creatinine or interfere with creatinine measurement assays (e.g., antibiotics, chemotherapeutic agents).

Quality assessment

Applicability and risk of study bias were assessed independently by 2 reviewers using the QUality Assessment of Diagnostic Accuracy Studies (QUADAS-2) [27]. The instrument includes “signaling” questions to assess bias in 4 domains (patient selection, index test, reference standard, and flow and timing) and questions in 3 domains (patient selection, index test, and reference standard) to assess the applicability of each study to the overall systematic review question.

In the patient selection domain, we assessed the appropriateness of the exclusion criteria relative to the aims of the specific study and against a pre-defined list of non-GFR determinants of creatinine; if exclusions were explained by these factors, we marked “yes” for the signaling question of whether the study avoided inappropriate exclusions. For pooled data studies, if it was unclear how the authors arrived at the number of participants compared with the original studies, we used a response of “unclear” for the exclusion criteria question. Concern that the included patients did not match the review question was rated as “high” if there was uncertainty that the study did not include only Black adults (e.g., participants <18 years old or of “mixed ancestry”). The index tests were defined as either the MDRD or CKD-EPI and the reference standard was mGFR using any of the following exogenous markers: 51Cr-EDTA, inulin, 125I-iothalamate, iohexol, or 99mTc-DTPA. In the flow and timing domain, an appropriate interval between the index and reference tests was defined as eGFR and mGFR samples drawn within 24 hours of each other.

If all responses to the signaling questions were “yes,” we assigned a rating of “low” to the domain; if all responses were “no,” we assigned a rating of “high.” For any combination of responses (i.e., any combination of “yes,” “no,” and/or “unclear”), we assigned a rating of “unclear.” Discordant ratings between reviewers were resolved through discussion.

Data synthesis. Studies were grouped by the methods used to assess eGFR vs. mGFR performance. Three categories of publications were identified: studies that evaluated eGFR equations 1) with and without race adjustment, 2) with race adjustment, and 3) without race adjustment. Data synthesis was conducted only on studies that assessed eGFR equations both with and without race adjustment compared to mGFR (category 1) in order to have directly comparable eGFR performance data for the review analysis. Additionally, when multiple publications reporting outcomes data on the same patient population were identified, the publication with the most complete data set was retained for analysis. Data are presented using descriptive tables. The outcomes of bias, accuracy, precision, and agreement measures (e.g., correlations and concordance), are summarized by eGFR equation. No minimum number of studies was needed to report results.

Results

In all, 24,850 results were retrieved (Fig 1) from database searches. Fifteen additional articles were identified through handsearching of 256 articles flagged for reference checking during the screening process. After removing duplicate records and pre-2015 conference abstracts, the titles and abstracts of 13,167 citations were screened. Of these, 11,919 were excluded after abstract review, and 1,190 were excluded at full-text review. Thirty-four articles met all eligibility criteria, of which, 12 met data synthesis criteria as described above.

thumbnail
Fig 1. Flow diagram of study search and selection process.

https://doi.org/10.1371/journal.pone.0276252.g001

Study characteristics

Twelve studies evaluating eGFR equations both with and without race adjustment vs. mGFR were included in the data synthesis (Table 1). See S3 Table for the study characteristics of the remaining 22 studies identified in the review.

thumbnail
Table 1. Summary characteristics of studies included in the systematic review synthesis.

https://doi.org/10.1371/journal.pone.0276252.t001

Of the 12 studies analyzed for the review that compared eGFR equations with and without race adjustment to mGFR, 7 (58.3%) were cross-sectional [10, 2833], 3 (25.0%) were retrospective [29, 33, 34], and 1 (8.3%) used pooled data from previous studies [10]; f4 (33.3%) were prospective [3538].

Participant clinical characteristics varied; 3 studies (25.0%) focused on participants with renal dysfunction [30, 36, 37] and 5 (41.6%) included a mixed population, including patients with and without kidney disease [10, 2829, 33, 38]. Participants with sickle cell disease [35] and HIV [3132, 34] were also represented.

All studies evaluating CKD-EPI used the 2009 creatinine-based equation (CKD-EPICr) [5]. Three of the studies that evaluated CKD-EPICr equations [30, 31, 34] also evaluated CKD-EPICr-Cys equations [39], with and without race correction (see S2 File). All studies evaluating MDRD used the 4-variable equation with a constant of 175 [3]. Measured GFR was assessed using iohexol in 4 studies [28, 32, 34, 35], 51Cr-EDTA in 4 studies [30, 31, 33, 36], 99mTc-DTPA in 2 studies [29, 38], and iothalamate in 2 studies [10, 37].

The geographic distribution of the studies varied. Three studies (25.0%) evaluated eGFR in the U.S. [10, 34, 37]. The U.S. studies included a retrospective validation study of HIV-positive and -negative individuals, which evaluated the bias and P30 of the CKD-EPICr equation [33], a cross-sectional validation study of patients with and without CKD, which evaluated the bias and root mean square error (RMSE) of the CKD-EPICr equation [10], and a prospective cohort study using data from the Chronic Renal Insufficiency Cohort (CRIC) study, which evaluated the bias of CKD-EPICr [37]. Six studies (50.0%) evaluated eGFR in African countries: South Africa [29, 31, 36, 38], the Ivory Coast [28], the Democratic Republic of the Congo [28], and Kenya [32]. One study (8.3%) was conducted in France [35], 1 study (8.3%) in the United Kingdom (UK) [33], and 1 (8.3%) was conducted in Brazil [30].

Table 2 presents the bias, accuracy, and precision performance results of the eGFR equations.

thumbnail
Table 2. Studies evaluating bias, accuracy, and precision, with and without race adjustment.

https://doi.org/10.1371/journal.pone.0276252.t002

Bias

Of the 11 studies evaluating bias of CKD-EPICr equations, with and without race adjustment, bias improved with removal of race adjustment in 10 studies (Table 2) [2835, 3738], including 2 U.S. studies [13, 37]. By contrast, a U.S. study of patients with and without CKD that used the original data set from which the CKD-EPICr equation was derived found that bias worsened with removal of race adjustment [10].

In all 7 studies evaluating bias of the 4-variable MDRD equation with and without race adjustment, bias improved with race adjustment removal [28, 3134, 36, 38].

Accuracy

All 8 studies, including one from the U.S. [33], evaluating the accuracy of CKD-EPICr equations, with and without race adjustment (Table 2), found improvement with removal of the race coefficient [2834, 38].

In all 6 studies assessing the accuracy of MDRD equations with and without race adjustment, removal of the race coefficient improved accuracy (Table 2) [28, 3133, 36, 38]. Across all studies reporting accuracy outcomes, irrespective of eGFR equation evaluated, all used P30 as an accuracy measure. Additional accuracy thresholds reported included P20 [38], P15 [31], and P10 [29, 32].

Precision

Precision improved with removal of race adjustment in 4 of 6 studies evaluating the precision of CKD-EPICr, with and without race adjustment (Table 2) [28, 31, 33, 38]. Precision was evaluated via different methodologies, including RMSE—calculated as the square root of the mean squared differences of eGFR and mGFR, the interquartile interval (IQR) of the difference between eGFR and mGFR, standard deviation of bias, or the interquartile range of the eGFR and mGFR differences. In 2 studies, including a U.S. study [10] and a study in a Brazilian population [30], precision worsened with the removal of race adjustment.

Five studies evaluated precision of the 4-variable MDRD equation (Table 2) and reported improvement when race adjustment was removed [28, 31, 33, 36, 38]. Precision was measured as standard deviation of bias or the interquartile range of the eGFR and mGFR differences.

Correlation and concordance

Overall, the findings were mixed in the 6 studies reporting correlation between CKD-EPICr equations, with and without race adjustment, and mGFR (S4 Table) [2830, 32, 35, 38]. Arlet and colleagues (2012) found that the difference between eGFR and mGFR decreased with increasing GFR values for CKD-EPICr with race adjustment (r = -0.23, p = 0.06) and without race adjustment (r = -0.43, p<0.001) [35]. Bukabau et al. report increased correlation between eGFR and mGFR in a large cohort of participants from the Democratic Republic of Congo and the Ivory Coast with removal of the race coefficient compared to inclusion of the race coefficient (Lin’s Concordance Correlation Coefficient of 0.81 vs. 0.71 respectively) [28]. Rocha and colleagues found that concordance was 52.5% for African Brazilians using CKD-EPICr with race adjustment, and 54.1% without race adjustment [30]. Moodley and colleagues [29] observed a high correlation both with and without race adjustment (R2 = 0.83 for females; R2 = 0.86 for males). Holness (2020) reports agreement between CKD-EPICr and mGFR; with race adjustment, the 95% limits of agreement were -14.5–55.2 and without race adjustment, -17.9–35.7 [38]. However, correlation was poor in a study by Wyatt and colleagues (R2 = 0.23 both with and without race adjustment) [32].

In 3 studies assessing the correlation between MDRD and mGFR (S4 Table), two reported increased correlation with removal of eGFR race adjustment [28, 35]. However, Wyatt and colleagues report no difference in correlation or variance inflation when the race variable is removed from the eGFR equation [32].

Social determinants of health and non-creatinine determinants of GFR

Only 2 studies reported social determinants of health and neither provided eGFR vs. mGFR performance outcomes (i.e., bias, accuracy, precision) by social determinants of health [34, 37]. Smoking status was reported in both studies, with Atta at al. also describing the income, education level, and insurance status of their study population (Black patients with mild to moderate CKD) [34]. Non-GFR determinants of creatinine were reported at baseline in 4 studies [32, 34, 35, 38]; however, none reported eGFR vs. mGFR performance by non-GFR determinants of creatinine.

Quality assessment outcomes

Three studies analyzed for the review had low risk of bias for the patient selection domain (Table 3). Nine studies had unclear risk of bias for patient selection (Table 3), including 3 retrospective studies [29, 33, 34] and 1 study evaluating outcomes based on multiple measurements for some patients [34]. All 12 studies analyzed for the review were rated as having unclear risk of bias for the index test and reference standard domains due to a lack of information about whether the tests were interpreted without knowledge of each other. Regarding flow and timing, 6 studies had low risk of bias [2830, 34, 36, 38], and the remaining had unclear bias. Applicability concerns were low for most studies and domains, except for patient selection in 1 study [38]. See S5 Table for quality assessment ratings of the 22 studies not included in the narrative synthesis.

thumbnail
Table 3. QUADAS-2 risk of bias and applicability assessment of studies included in the systematic review synthesis.

https://doi.org/10.1371/journal.pone.0276252.t003

Discussion

Previous efforts revealed the need for a comprehensive, systematic review of international scope to inform the use of eGFR estimation in Black patients [40], and our study is among the first systematic reviews to assess worldwide evidence related to adjustment for Black race in eGFR equations. The most commonly reported performance metrics identified from our systematic review were bias, precision, and accuracy, which improved in the majority of studies when race was removed from eGFR equations. Concordance and correlation were less often evaluated and seldom assessed in a standardized way, making it difficult to draw conclusions. Results from the QUADAS-2 quality assessment indicated that while there was low concern regarding the applicability of the included studies for the overall systematic review question, risk of study bias was difficult to assess due to inadequate reporting and variations in study methodology and design.

Recently, new recommendations have been put forth by NKF-ASN and the National Institute for Health and Care Excellence (NICE) to remove race adjustment from eGFR equations [20, 21, 41, 42]. The NKF-ASN task force recommended a new CKD-EPICr equation refit without a race adjustment for U.S. adults [20, 21]. We did not include the study reporting the new CKD-EPICr equation [21] in our systematic review, since it was published outside of our pre-established date parameters. While the NKF-ASN task force recommended use of the new CKD-EPICr equation refit without race, they noted that accuracy may improve with use of equations combining filtration markers. Inker and colleagues (2021) found that use of a new CKD-EPI equation incorporating both creatinine and cystatin C, without race adjustment (new CKD-EPICr-Cys), was more accurate than the new CKD-EPICr equation, which underestimated mGFR in Black adults and overestimated mGFR in non-Black adults in the U.S. [21]. Future validation studies are needed to assess the performance of the new CKD-EPI equations in other countries and across a variety of clinical settings.

Our study has several limitations. We only included studies that compared the performance of eGFR equations both with and without race adjustment. We did not evaluate differences in performance between the multiple versions of the CKD-EPI and MDRD equations, thought less clinically relevant especially given recent guidelines. Measured GFR was not assessed consistently between studies. Many of the included studies did not assess the statistical significance of outcome measures when race adjustment was removed. None of the included studies assessed the impact of social determinants of health and non-creatinine determinants of GFR. Many studies did not report how race was determined; since race is inconsistently defined across populations [43], this contributes to heterogeneity in the findings. Risk of overall study bias was unclear for many studies due to inadequate reporting and variations in study methodology. A further limitation is the potential risk of publication bias; however, we minimized this risk by conducting a comprehensive literature review that spanned across multiple databases and included grey literature sources.

Our study focused on Black participants rather than mixed populations. While we recognize the value in studies with mixed populations, we chose to focus on studies with Black participants in order to optimally assess the study’s main objective of determining how well eGFR with and without race adjustment estimates mGFR in Black adults across the world. Given the paucity of data on eGFR in Black people in the U.S., and the historical limited participation of Black people in clinical trials including kidney-focused trials [44], the focus of this review on Black participants is an added strength of our study.

Limitations in eGFR due to variation in non-GFR determinants of serum creatinine could be relevant in our systematic review, which included studies with heterogenous populations. In studies of sicker patient populations, eGFRCr likely overestimates kidney function, and removal of the race coefficient can bring kidney function closer to the expected value. However, such findings are coincidental and do not provide additional evidence to support that eGFR equations without the race coefficient outperform eGFR equations with the race coefficient. The sub-optimal accuracy measures observed in many of the studies included in our systematic review support the need for continued efforts to improve the accuracy of eGFR within and outside the U.S. More modern and elaborate approaches are gradually replacing existing simplistic methods used in MDRD and CKD-EPI equations. Examples include Q-values in the new EKFC-equation [45] and creatinine growth curves [46]. Widespread use of these elaborate methods is currently limited by a paucity of external validation studies.

This novel, in-depth and worldwide systematic review, provides the highest level of evidence against race adjustment when estimating GFR [20, 21, 41, 42]. Race is an inappropriate proxy for genetics all over the world, and efforts to eliminate its use in estimating GFR should be global. In addition to tracking the performance of the newly recommended race-free eGFR equations, future research should prioritize health equity [47, 48] by assessing the impact of social determinants of racial disparities in kidney disease on GFR estimates and ensuring enrollment of diverse participants across the world in validation trials of GFR equations [47, 48].

Supporting information

S1 File. Search strategy and processes details.

https://doi.org/10.1371/journal.pone.0276252.s001

(DOCX)

S2 File. Studies evaluating CKD-EPICr-Cys equations with and without race adjustment.

https://doi.org/10.1371/journal.pone.0276252.s002

(DOCX)

S3 Table. Summary characteristics of studies not included in the systematic review synthesis.

https://doi.org/10.1371/journal.pone.0276252.s005

(DOCX)

S4 Table. Correlation or concordance tables for CKD-EPICr and MDRD studies with and without race adjustment.

https://doi.org/10.1371/journal.pone.0276252.s006

(DOCX)

S5 Table. QUADAS-2 risk of bias and applicability assessment of studies not included in the systematic review synthesis.

https://doi.org/10.1371/journal.pone.0276252.s007

(DOCX)

References

  1. 1. Stevens LA, Levey AS. Measured GFR as a confirmatory test for estimated GFR. J Am Soc Nephrol JASN. 2009;20(11):2305–2313. pmid:19833901
  2. 2. Shahbaz H, Gupta M. Creatinine Clearance. In: StatPearls. StatPearls Publishing; 2021. Accessed December 9, 2021. http://www.ncbi.nlm.nih.gov/books/NBK544228/
  3. 3. Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Modification of Diet in Renal Disease Study Group. Ann Intern Med. 1999;130(6):461–470. pmid:10075613
  4. 4. Stevens PE, Levin A; Kidney Disease: Improving Global Outcomes Chronic Kidney Disease Guideline Development Work Group Members. Evaluation and management of chronic kidney disease: synopsis of the Kidney Disease: Improving Global Outcomes 2012 clinical practice guideline. Ann Intern Med. 2013;158(11):825–830. pmid:23732715
  5. 5. Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro AF 3rd, Feldman HI, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604–612. pmid:19414839
  6. 6. Peralta CA, Lin F, Shlipak MG, Siscovick D, Lewis C, Jacobs DR Jr, et al. Race differences in prevalence of chronic kidney disease among young adults using creatinine-based glomerular filtration rate-estimating equations. Nephrol Dial Transplant. 2010;25(12):3934–3939. pmid:20519233
  7. 7. Burchard EG, Ziv E, Coyle N, Gomez SL, Tang H, Karter AJ, et al. The importance of race and ethnic background in biomedical research and clinical practice. N Engl J Med. 2003;348(12):1170–1175. pmid:12646676
  8. 8. Vyas DA, Eisenstein LG, Jones DS. Hidden in plain sight—reconsidering the use of race correction in clinical algorithms. N Engl J Med. 2020;383(9):874–882. pmid:32853499
  9. 9. Morris H, Mohan S. Using race in the estimation of glomerular filtration rates: time for a reversal? Curr Opin Nephrol Hypertens. 2020;29(2):227–231. pmid:31895163
  10. 10. Levey AS, Tighiouart H, Titan SM, Inker LA. Estimation of glomerular filtration rate with vs without including patient race. JAMA Intern Med. 2020;180(5):793–795. pmid:32176270
  11. 11. Eneanya ND, Yang W, Reese PP. Reconsidering the consequences of using race to estimate kidney function. JAMA. 2019;322(2):113–114. pmid:31169890
  12. 12. Yudell M, Roberts D, DeSalle R, Tishkoff S. Science and society. Taking race out of human genetics. Science. 2016;351(6273):564–565. pmid:26912690
  13. 13. Institute of Medicine (US) Committee on Understanding and Eliminating Racial and Ethnic Disparities in Health Care. Unequal Treatment: Confronting Racial and Ethnic Disparities in Health Care. (Smedley BD, Stith AY, Nelson AR, eds.). National Academies Press (US); 2003. Accessed December 9, 2021. http://www.ncbi.nlm.nih.gov/books/NBK220358/
  14. 14. Eneanya ND, Boulware LE, Tsai J, Bruce MA, Ford CL, Harris C, et al. Health inequities and the inappropriate use of race in nephrology. Nat Rev Nephrol. 2021;1–11.
  15. 15. Centers for Disease Control and Prevention. Chronic kidney disease in the United States, 2021. Published March 4, 2021. Accessed December 9, 2021. https://www.cdc.gov/kidneydisease/publications-resources/ckd-national-facts.html
  16. 16. National Institute for Diabetes and Digestive and Kidney Diseases. Kidney disease statistics for the United States. Published September 2021. Accessed December 9, 2021. https://www.niddk.nih.gov/health-information/health-statistics/kidney-disease
  17. 17. Williams WW, Delmonico FL. The end of racial disparities in kidney transplantation? Not so fast! J Am Soc Nephrol. 2016;27(8):2224–2226. pmid:26952001
  18. 18. Powe NR. Black kidney function matters: use or misuse of race? JAMA. 2020;324(8):737–738. pmid:32761164
  19. 19. National Kidney Foundation, American Society of Nephrology. Establishing a task force to reassess the inclusion of race in diagnosing kidney diseases. National Kidney Foundation. Published July 2, 2020. Accessed December 9, 2021. https://www.kidney.org/news/establishing-task-force-to-reassess-inclusion-race-diagnosing-kidney-diseases
  20. 20. Delgado C, Baweja M, Crews DC, et al. A unifying approach for GFR estimation: recommendations of the NKF-ASN Task Force on reassessing the inclusion of race in diagnosing kidney disease. Am J Kidney Dis. Published online September 22, 2021:S0272-6386(21)00828-3. pmid:34563581
  21. 21. Inker LA, Eneanya ND, Coresh J, Tighiouart H, Wang D, Crews DC, et al. New creatinine- and cystatin C-based equations to estimate GFR without race. N Engl J Med. 2021;385(19):1737–1749. pmid:34554658
  22. 22. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. PLoS Med. 2021;18(3):e1003583. pmid:33780438
  23. 23. Rethlefsen ML, Kirtley S, Waffenschmidt S, Ayala AP, Moher D, Page MJ, et al. PRISMA-S: an extension to the PRISMA Statement for Reporting Literature Searches in Systematic Reviews. Syst Rev. 2021;10(1):39. pmid:33499930
  24. 24. Shamseer L, Moher D, Clarke M, Ghersi D, Liberati A, Petticrew M, et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015: elaboration and explanation. BMJ. 2015;350:g7647. pmid:25555855
  25. 25. Harzing AW. Publish or Perish [computer program]. Version 7. 2007. https://harzing.com/resources/publish-or-perish
  26. 26. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42(2):377–381. pmid:18929686
  27. 27. Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, et al. QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med. 2011;155(8):529–536. pmid:22007046
  28. 28. Bukabau JB, Yayo E, Gnionsahé A, Monnet D, Pottel H, Cavalier E, et al. Performance of creatinine- or cystatin C-based equations to estimate glomerular filtration rate in sub-Saharan African populations. Kidney Int. 2019 May;95(5):1181–1189. pmid:30910379
  29. 29. Moodley N, Hariparshad S, Peer F, Gounden V. Evaluation of the CKD-EPI creatinine based glomerular filtration rate estimating equation in Black African and Indian adults in KwaZulu-Natal, South Africa. Clin Biochem. 2018;59:43–49. pmid:29940141
  30. 30. Rocha AD, Garcia S, Santos AB, Eduardo JCC, Mesquita CT, Lugon JR, et al. No race-ethnicity adjustment in CKD-EPI equations is required for estimating glomerular filtration rate in the Brazilian population. Int J Nephrol. 2020;2020:2141038. pmid:32733708
  31. 31. Seape T, Gounden V, van Deventer HE, Candy GP, George JA. Cystatin C- and creatinine-based equations in the assessment of renal function in HIV-positive patients prior to commencing Highly Active Antiretroviral Therapy. Ann Clin Biochem. 2016;53(Pt 1):58–66. pmid:25766385
  32. 32. Wyatt CM, Schwartz GJ, Owino Ong’or W, Abuya J, Abraham AG, Mboku C, et al. Estimating kidney function in HIV-infected adults in Kenya: comparison to a direct measure of glomerular filtration rate by iohexol clearance. PloS One. 2013;8(8):e69601. pmid:23950899
  33. 33. Gama RM, Clery A, Griffiths K, Heraghty N, Peters AM, Palmer K, et al. Estimated glomerular filtration rate equations in people of self-reported Black ethnicity in the United Kingdom: inappropriate adjustment for ethnicity may lead to reduced access to care. PloS One. 2021;16(8):e0255869. pmid:34383841
  34. 34. Atta MG, Zook K, Brown TT, Vaidya D, Tao X, Maier P, et al. Racial adjustment adversely affects glomerular filtration estimates in Black Americans living with HIV. J Am Soc Nephrol. 2021;32(9):2143–2147. pmid:34140402
  35. 35. Arlet JB, Ribeil JA, Chatellier G, Eladari D, De Seigneux S, Souberbielle JC, et al. Determination of the best method to estimate glomerular filtration rate from serum creatinine in adult patients with sickle cell disease: a prospective observational cohort study. BMC Nephrol. 2012;13:83. pmid:22866669
  36. 36. van Deventer HE, George JA, Paiker JE, Becker PJ, Katz IJ. Estimating glomerular filtration rate in Black South Africans by use of the Modification of Diet in Renal Disease and Cockcroft-Gault equations. Clin Chem. 2008;54(7):1197–1202. pmid:18487286
  37. 37. Zelnick LR, Leca N, Young B, Bansal N. Association of the estimated glomerular filtration rate with vs without a coefficient for race with time to eligibility for kidney transplant. JAMA Netw Open. 2021;4(1):e2034004. pmid:33443583
  38. 38. Holness JL, Bezuidenhout K, Davids MR, Warwick JM. Validation of equations to estimate glomerular filtration rate in South Africans of mixed ancestry. South Afr Med J. 2020;110(3):229–234. pmid:32657701
  39. 39. Inker LA, Schmid CH, Tighiouart H, Eckfeldt JH, Feldman HI, et al. Estimating glomerular filtration rate from serum creatinine and cystatin C. N Engl J Med. 2012;367(1):20–29. pmid:22762315
  40. 40. Whitney K. Group’s efforts lead to removal of race as a variable in common test of kidney function. VUMC Reporter. Published July 13, 2020. Accessed December 9, 2021. https://news.vumc.org/2020/07/13/groups-efforts-lead-to-removal-of-race-as-a-variable-in-common-test-of-kidney-function/
  41. 41. National Institute for Health and Care Excellence (NICE). Chronic kidney disease: assessment and management. NICE. Published August 25, 2021. Accessed December 14, 2021. https://www.nice.org.uk/guidance/NG203
  42. 42. NICE Guideline Updates Team. Evidence reviews for the diagnostic accuracy of eGFR calculations in adults, children, and young people from black, Asian and other minority ethnic groups with CKD: Chronic kidney disease: Evidence review A. London: National Institute for Health and Care Excellence (NICE); 2021 Aug. 200 p. Available from: https://www.nice.org.uk/guidance/ng203/evidence/a-diagnostic-accuracy-of-egfr-calculations-in-adults-children-and-young-people-from-black-asianand-other-minority-ethnic-groups-with-ckd-pdf-9204445982
  43. 43. Shanawani H, Dame L, Schwartz DA, Cook-Deegan R. Non-reporting and inconsistent reporting of race and ethnicity in articles that claim associations among genotype, outcome, and race or ethnicity. J Med Ethics. 2006 Dec;32(12):724–8. pmid:17145914
  44. 44. Gadegbeku CA, Stillman PK, Huffman MD, Jackson JS, Kusek JW, Jamerson KA. Factors associated with enrollment of African Americans into a clinical trial: results from the African American study of kidney disease and hypertension. Contemp Clin Trials. 2008 Nov;29(6):837–42. pmid:18639652
  45. 45. Pottel H, Björk J, Courbebaisse M, Couzi L, Ebert N, Eriksen BO, et al. Development and Validation of a Modified Full Age Spectrum Creatinine-Based Equation to Estimate Glomerular Filtration Rate: A Cross-sectional Analysis of Pooled Data. Ann Intern Med. 2021 Feb;174(2):183–191. pmid:33166224
  46. 46. Björk J, Nyman U, Larsson A, Delanaye P, Pottel H. Estimation of the glomerular filtration rate in children and young adults by means of the CKD-EPI equation with age-adjusted creatinine values. Kidney Int. 2021 Apr;99(4):940–947. pmid:33157151
  47. 47. Artiga S, Hinton E. Beyond health care: the role of social determinants in promoting health and health equity. Kaiser Family Foundation. Published May 10, 2018. Accessed December 17, 2021. https://www.kff.org/racial-equity-and-health-policy/issue-brief/beyond-health-care-the-role-of-social-determinants-in-promoting-health-and-health-equity/
  48. 48. Norton JM, Moxey-Mims MM, Eggers PW, Narva AS, Star RA, Kimmel PL, et al. Social determinants of racial disparities in CKD. J Am Soc Nephrol. 2016 Sep;27(9):2576–95. pmid:27178804