Next Article in Journal
Pancreatic Adeno-MiNEN, a Rare Newly Defined Entity with Challenging Diagnosis and Treatment: A Case Report with Systematic Literature Review and Pooled Analysis
Next Article in Special Issue
Renal Tissue Expression of BAFF and BAFF Receptors Is Associated with Proliferative Lupus Nephritis
Previous Article in Journal
A Real-Life Action toward the End of HIV Pandemic: Surveillance of Mother-to-Child HIV Transmission in a Center from Southeast Romania
Previous Article in Special Issue
Progress in the Pathogenesis and Treatment of Neuropsychiatric Systemic Lupus Erythematosus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Predictors of Early Response, Flares, and Long-Term Adverse Renal Outcomes in Proliferative Lupus Nephritis: A 100-Month Median Follow-Up of an Inception Cohort

1
Department of Nephrology and Renal Transplantation, Laiko Hospital, Medical School, National & Kapodistrian University of Athens, 11527 Athens, Greece
2
Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National & Kapodistrian University of Athens, 11527 Athens, Greece
3
Department of Pathology, Medical School, National & Kapodistrian University of Athens, 11527 Athens, Greece
4
Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Laiko Hospital, Medical School, National & Kapodistrian University of Athens, 11527 Athens, Greece
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(17), 5017; https://doi.org/10.3390/jcm11175017
Submission received: 27 July 2022 / Revised: 20 August 2022 / Accepted: 22 August 2022 / Published: 26 August 2022
(This article belongs to the Special Issue Systemic Lupus Erythematosus: Pathogenesis, Diagnosis and Treatment)

Abstract

:
Objective: To define predictors of response, time to response, flares, and long-term renal outcome in an inception cohort of proliferative lupus nephritis (PLN). Methods: We included 100 patients (80% female; mean age 31 ± 13 years) with biopsy-proven PLN (III, IV, III/IV + V). Clinical, laboratory, histological and therapeutical parameters were recorded at baseline, 6, 9, 12, 18, 24, 36, 72 months, time of flare, and last follow-up visit. Logistic and Cox-regression models were applied. Results: After induction treatment (69% received cyclophosphamide (CYC) and 27% mycophenolic acid (MPA)), partial (PR) or complete (CR) response was achieved in 59% (26% CR, 33% PR) and 67% (43% CR, 24% PR) of patients at 3 and 6 months, respectively; median time to PR was 3 months (IQR 5) and median time to CR was 6 months (IQR 9). Baseline proteinuria <1.5 g/day correlated with a shorter time to CR (HR 1.77) and with CR at 3, 6, and 9 months (OR 9.4, OR 5.3 and OR 3.7, respectively). During 100-month median follow-up, 33% of patients had ≥1 renal flares (median time: 38 months). Proteinuria >0.8 g/day at 12 months was associated with a higher risk of flares (OR 4.12), while MPA and mixed classes with lower risk (OR 0.14 and OR 0.13, respectively). Baseline proteinuria >2 g/day and 12-month proteinuria >0.8 g/day correlated with a shorter time to flare (HR 2.56 and HR 2.57, respectively). At the end of follow-up, 10% developed stage 3–4 chronic kidney disease (CKD), and 12% end-stage renal disease (ESRD). Twelve-month proteinuria >0.8 g/day (OR 10.8) and interstitial fibrosis/tubular atrophy >25% (OR 7.7) predicted CKD or ESRD at last visit. Conclusions: Baseline proteinuria <1.5 g/day predicted time to CR. Twelve-month proteinuria >0.8 g/day correlated with flares (ever) and time to flare and, along with baseline interstitial fibrosis/tubular atrophy >25%, predicted CKD or ESRD at the last visit.

1. Introduction

In systemic lupus erythematosus (SLE), renal involvement is the most common severe complication of the disease affecting approximately 20–60% of patients [1] and conferring a high morbidity and mortality risk [2,3,4]. End-stage renal disease (ESRD) still occurs at varying rates among different cohorts of patients with lupus nephritis (LN), despite the revolutionary changes in LN treatment over the past fifty years [5,6]. A wide range of demographic, socioeconomic and disease-related parameters have been associated with LN’s short- and long-term outcomes. Histological class is a major determinant of renal survival in LN, with the proliferative classes having the worst prognosis compared to other classes [5,7].
The main therapeutic goal in patients with LN is the long-term preservation of renal function, preventing and managing comorbidities, and improving disease-related quality of life [8]. These goals can be achieved by inducing and maintaining disease remission and by preventing and timely treating disease flares [9,10]. In this context, recognizing early predictors of renal response, flare and long-term renal survival is paramount in guiding the therapeutic management of LN patients.
The aim of the present study is (a) to examine the response to treatment and short- and long-term renal outcomes in an inception cohort of patients with proliferative LN; (b) to define predictors of response, time to response, flares and long-term adverse renal outcomes.

2. Patients and Methods

2.1. Study Population

We examined an inception cohort of 100 patients with proliferative LN diagnosed between 1992 and 2021 and followed up at our joint academic center (Nephrology and Rheumatology Units) at Laiko General Hospital of Athens until April 2022. All patients fulfilled the 2019 classification criteria for SLE and had a biopsy-proven diagnosis of proliferative LN (class III, IV, III/IV + V), classified according to the International Society of Nephrology/ Renal Pathology Society (ISN/RPS) 2003 lupus nephritis classification system. Among 100 patients identified with PLN, 28% (28/100) had class III, 47% (47/100) class IV, 9% (9/100) mixed class III + V and 16% (16/100) mixed class IV + V.

2.2. Data Collection

We retrospectively reviewed medical charts of patients and recorded clinical, laboratory, histological and therapeutical parameters at the following time points: time of histological diagnosis of LN; 3, 6, 9, 12, 18, 24, 36, 72 months after the diagnosis; time of renal flare (with or without a repeat biopsy), and at last follow-up visit. Patients with inadequate data and less than 6 months of follow-up were excluded.
Data collected included demographic parameters, time from SLE diagnosis to LN onset, disease activity (assessed by Systemic Lupus Erythematosus Disease Activity Index 2000, SLEDAI-2K score) [11], anti-ds DNA titers, C3 and C4 levels, serum urea (Ur), creatinine (Cr) and albumin, eGFR (based on the CKD-EPI formula), 24-h proteinuria, urine sediment, renal biopsy histological parameters (LN class, activity index (AI), chronicity index (CI), crescents, interstitial fibrosis/tubular atrophy (IF/TA)), and treatment regimens.
All data (demographic, clinical, laboratory and histological) were extracted from our anonymized cohort dataset. No individualized or identifiable data are presented in this study. The study was approved by the Institutional Review Board (protocol number 1725/14-12-2017) of Laiko General Hospital of Athens.

2.3. Patient and Public Involvement

Patients and/or the public were not involved in this research’s design, conduct, reporting or dissemination plans.

2.4. Definitions

Response and flares were defined according to the 2012 EULAR/ERA-EDTA [8] and the 2012 KDIGO recommendations [12] as follows: Active urine sediment: the presence of >5 RBCs/hpf or ≥1 red cell casts. Complete response (CR): proteinuria <500 mg/24 h and serum creatinine within 10% of baseline values. Partial response (PR): ≥50% reduction in proteinuria to subnephrotic levels and serum creatinine within 10% of baseline values. Nephritic flare: increase in glomerular haematuria by ≥10 RBCs/hpf with or without a decrease in eGFR by ≥10%, irrespective of changes in proteinuria. Nephrotic flare: reproducible doubling of proteinuria to >1000 mg/24 h if a complete response had been previously achieved or reproducible doubling of proteinuria to ≥2000 mg/24 h if a partial response has been previously achieved. Stage 3–4 chronic kidney disease (CKD): eGFR 15–60 mL/min/1.73 m2 for more than 3 months, and end-stage renal disease (ESRD) as eGFR < 15 mL/min/1.73 m2 or initiation of renal replacement therapy [13].

2.5. Statistical Analysis

Continuous variables were expressed as the mean value and standard deviation or median value and interquartile range (IQR), whereas categorical variables as frequencies and percentages. To investigate the differences in baseline parameters between patients with different therapeutical schemes, the t-test and Mann–Whitney U test for independent samples for continuous variables and the χ2 and Fisher exact test for categorical variables were applied. Univariate logistic regression analyses were performed to estimate the prognostic effect of various variables on the risk of renal flare and adverse renal outcomes. Cox regression analyses investigated the association between patients’ variables and time to response or response at specific time points. Variables found to be significant in the univariate analyses were included in the multivariate models. In the models identifying predictors of flare and adverse renal outcome, due to a small number of events, selected independent variables were included in the multivariate analyses after multicollinearity was examined. Multicollinearity issues were assessed with appropriate tests (χ2, Cramer’s V coefficient (V), analysis of variants (ANOVA), Spearman correlation coefficient (rs) and Mann–Whitney U test) before performing multivariate analyses. Strong correlation among specific variables excluded some parameters from the multivariate models. Significance was set at α = 0.05. The estimated odds ratios (ORs) and hazard ratios (HRs) of both the univariate and multivariate models and the related p-values are presented. Data were analyzed using Stata 13.0 software (Stata Corporation, College Station, TX). All tests proceeded as two-tailed.

3. Results

3.1. Baseline Characteristics

We studied 100 patients with biopsy-proven proliferative LN. The baseline demographic, clinical, laboratory and histological characteristics and the therapeutic regimens applied are shown in Table 1. All biopsies contained ≥10 glomeruli, and those performed before 2003 were reassessed based on ISN/RPS 2003 classification system. The median follow-up time was 100 months (IQR = 108).

3.2. Immunosuppressive Regimens

Induction treatment consisted of cyclophosphamide (CYC) in 69% (69/100) of patients (in combination with rituximab (RTX) in 12/69), mycophenolic acid (MPA) in 27% (27/100, in combination with RTX in 4/27) and RTX alone in 2 patients. All the above patients received corticosteroids. One patient was treated only with corticosteroids due to non-compliance, while one patient did not receive any immunosuppressive treatment due to progression to ESRD soon after LN diagnosis (Table 1). CYC was given in all 69 patients intravenously, following the NIH regimen in 96% and the ELNT regimen in 4% of cases.
Patients treated with CYC differed significantly in several baseline parameters compared to those treated with MPA (Supplemental Table S1). Although baseline eGFR did not differ between the two treatment groups, all patients with eGFR <30 mL/min/1.73 m2 were treated with CYC. Half of the patients with class III were treated with CYC and half with MPA (13/26 vs. 13/26). CYC was more often preferred over MPA for patients with class IV (87% (40/46) vs. 13% (6/46)) and for patients with class IV + V (88% (14/16) versus 12% (2/16)). Patients with class III + V received more often MPA than CYC (75% (6/8) versus 25% (2/8)) (data not shown).
Maintenance treatment consisted of MPA in 77% (77/100) of patients, azathioprine (AZA) in 8% (8/100) and CYC in 5% (5/100). Ten patients (10/100, 10%) did not receive any immunosuppressive maintenance treatment (Table 1); 5/10 due to early progression to ESRD, 2/10 who received RTX as induction continued with steroids only, 1/10 due to non-compliance, and 2/10 were lost to follow-up after completing the 6-month induction treatment.
The median duration of treatment was 39 months (IQR = 38) and did not differ between the CYC and MPA groups. Additionally, 76% (76/100) of patients received hydroxychloroquine and 65% (65/100) an ACEi or ARB.

3.3. Renal Response and Determinants of Response

For up to 72 months, 59% of patients achieved complete (CR) or partial (PR) response at 3 months (26% CR, 33% PR), 67% at 6 months (43% CR, 24% PR), 75% at 9 months (45% CR, 30% PR), 88% at 12 months (69% CR, 19% PR), and 86% at 18 months (71% CR, 15% PR), with similar percentages at all the following time points (Figure 1).
The median time to PR and CR was 3 months (IQR = 5) and 6 months (IQR = 9), respectively. Time to CR was significantly shorter in patients with class III (median/IQR 3/4) compared to patients with class IV (median/IQR 10/13, p <0.001) and those with mixed classes (median/IQR 10/11, p = 0.006) in the 6, 9, 12 and 18-month time points (p for log-rank = 0.49, p for Wilcoxon = 0.02) (Supplemental Figure S1). There was no difference in time to CR between class IV and mixed classes (p = 0.94). Time to response between the two treatment groups (CYC versus MPA) did not differ significantly (Supplemental Figure S1). Median time to CR was 6 months (IQR = 7.5) in the MPA group and 7 months (IQR = 15) in the CYC group (p = 0.09).
Moreover, 7% of patients (7/100) of the cohort did not ever respond to treatment, 6 of whom progressed to ESRD, while one patient had a preserved renal function at 7 months but was lost to follow-up afterward. Two nonresponders, who progressed to ESRD after 4 and 5 months, died at 6 and 7 months, respectively, after LN diagnosis.
In Cox regression analysis, proteinuria <1.5 g/day at LN diagnosis was the only parameter that significantly correlated with a shorter time to complete response (HR 1.77, p = 0.01) (Table 2).The two treatment groups were compared when they were used alone or in combination with RTX.
We further compared patients who achieved CR at 12-18-24 months versus those with PR or no response at the respective time points. We found that the only significant predictor of CR was baseline proteinuria <1.5 g/day (OR 16.9, p = 0.008 for month 12, OR 5.24, p = 0.01 for month 18 and OR 4, p = 0.03 for month 24) (Supplemental Table S2). Although achievement of CR in earlier time points was less frequent, baseline proteinuria <1.5 g/day was also found to be the only significant predictor of CR at 3 months (OR 9.4, p <0.001) and 6 months (OR 5.3, p = 0.004), and, along with baseline eGFR > 60 mL/min (OR 4.04, p = 0.02), predicted CR at 9 months (OR 3.7, p = 0.02) (Supplemental Table S2).

3.4. Renal Flares and Determinants of Flares

Thirty-three percent (33/100) of patients had ≥1 renal flares in a median time of 38 months (IQR = 43); 30% (10/33) of flares were nephritic, and 70% (23/33) nephrotic. Repeat biopsy was performed in 91% (30/33) of cases, and a class switch was observed in 67% (20/30) of them. In all 30 repeat biopsies, active LN lesions were confirmed. In two of them, a pattern of focal segmental glomerulosclerosis was also described, which may have contributed to the nephrotic clinical presentation of these patients.
In univariate logistic regression analysis, longer time to response was identified as a significant risk factor of flare (OR 1.14, p = 0.01 for PR, OR 1.05, p = 0.01 for CR) (Table 3). Response (CR or PR) at 3 months (OR 1.05, p = 0.9), 6 months (OR 1.25, p = 0.66) and 9 months (OR 2.2, p = 0.19) after treatment did not significantly affect the risk of flare. Statistical significance was found for lack of response (CR or PR) at 12 (OR 3.8, p = 0.04), 18 (OR 4.9 p = 0.01) and 24 (OR 6.6, p = 0.02) months (Supplemental Table S3).
Proteinuria >2 g/day at diagnosis (OR 3, p = 0.02), and proteinuria >0.8 g/day at 12 months (OR 3.38, p = 0.02) were significantly associated with a higher risk of flare. Conversely, parameters associated with a lower risk of flares were older age (OR 0.95, p = 0.02) and treatment with MPA (OR 0.25, p = 0.02) compared to treatment with CYC (whether these agents were used alone or in combination with RTX). Mixed classes were also found to reduce the risk of flare (OR 0.21, p = 0.02), but this association was significant only when mixed classes were compared to class III and not to class IV (Table 3).
Since proteinuria at diagnosis, as a variable, was found to be strongly associated with induction therapy, histological class, and proteinuria at 12 months, it was not included in the multivariate model for the risk of flare. Accordingly, time to remission was strongly correlated with proteinuria at 12 months and LN class and was not included in the multivariate model (Supplemental Table S4).
In the multivariate analysis, proteinuria >0.8 g/day at 12 months was a significant predictor of flare (OR 4.12, p = 0.02), while treatment with MPA (OR 0.14, p = 0.01) and mixed classes were associated with lower flare risk (OR 0.13, p = 0.02) (the latter only when compared to class III) (Table 3).
Baseline proteinuria >2 g/day (HR 2.56, p = 0.02) and proteinuria >0.8 g/day at 12 months (HR 2.57, p = 0.01) were also found to correlate with shorter time to flare (Figure 2).

3.5. Patient Survival

At a median follow-up time of 100 months, four deaths were reported. A 59-year-old and heavy smoker patient died due to lung cancer 8 years after LN diagnosis, during which he was on immunosuppressive treatment (he received CYC as induction treatment followed by MPA for 3 years and AZA, due mainly to extrarenal manifestations, for 5 years). One patient, aged 65, died due to bladder cancer 8 years after LN diagnosis. Interestingly, this patient had never received CYC. Two other patients, aged 30 and 38, died from sepsis (7 and 6 months after LN diagnosis, respectively). Both patients had presented with rapid deterioration of renal function and progressed to ESRD very soon after LN diagnosis.

3.6. Renal Survival

At the end of the follow-up period, 10% (10/100) of patients developed stage 3–4 CKD, and 12% (12/100) progressed to ESRD.
Six of 12 (50%) patients who reached ESRD were nonresponders, and in 5 of them, ESRD was developed in the first 9 months. Seven (58%) patients progressed to ESRD after 55–183 months from LN diagnosis. eGFR at the time of diagnosis, renal response, and time until ESRD are shown in Supplemental Table S5.
We defined the composite endpoint of stage 3–4 CKD and ESRD as adverse long-term renal outcomes. Of note, all patients who had a decrease of eGFR >25% of baseline values at the last follow-up had reached stage 3–4 CKD or ESRD.
In the univariate logistic regression analysis, adverse long-term renal outcome was significantly associated with baseline eGFR < 60 mL/min/1.73 m2 (OR 6, p = 0.001), interstitial fibrosis/tubular atrophy >25% (OR 5.44, p = 0.007), proteinuria >0.8 g/day at 12 months (OR 9.5, p = 0.003) and longer time to response (OR 1.1, p = 0.01) (Table 4). The risk for stage 3–4 CKD or ESRD was greater for those not achieving any response after 6 months of treatment (OR 6.4, p = 0.001), while response at 3 months did not seem to affect long-term renal outcome significantly (OR 2.2, p = 0.11) (Supplemental Table S6).
eGFR at diagnosis was strongly correlated with baseline proteinuria, proteinuria at 12 months, and interstitial fibrosis/tubular atrophy. Therefore, it was excluded from the multivariate model. Time to remission was strongly correlated with baseline proteinuria, proteinuria at 12 months, and eGFR at diagnosis and was not included in the multivariate model either (Supplemental Table S7).
In multivariate analysis, proteinuria >0.8 g/day at 12 months (OR 10.8, p = 0.001) and interstitial fibrosis/tubular atrophy >25% at LN diagnosis (OR 7.7, p = 0.01) remained significant predictors of adverse renal outcome (Table 4).

4. Discussion

A major therapeutic goal in patients with LN is the long-term preservation of renal function by minimizing chronic active or relapsing disease and, at the same time, avoiding excessive drug toxicity. In this context, we need to identify predictors of renal response, relapse, and long-term adverse renal outcomes to optimize our therapeutic strategies. Short-term predictors can also serve as endpoints in large clinical trials investigating new therapeutic agents [14,15,16,17]. In our study, baseline proteinuria <1.5 g/day was the only significant predictor of time to complete response, while proteinuria >0.8 g/day at 12 months and treatment with CYC (compared to MPA) emerged as strong predictors of renal flare. We also showed that adverse long-term renal outcomes (stage 3–4 CKD, ESRD) could only be predicted by interstitial fibrosis/tubular atrophy >25% in baseline biopsy and proteinuria >0.8 g/day at 12 months.
In our study, response to treatment was achieved early after induction treatment; 59% of patients had a renal response at 3 months (26% CR, 33% PR), 67% at 6 months (43% CR, 24% PR), 88% at 12 months (69%CR, 19% PR) and 88% (71% CR, 17% PR) at 2 years. These results are encouraging compared to previous studies reporting 33–50% CR at 6 months, 49–68% CR at 12 months, and 63% CR at 2 years [9,14,18,19,20,21,22,23,24], although our population was predominantly Caucasian. Response to treatment has been associated with better long-term renal outcomes [25,26,27,28], with some studies reporting better renal survival in patients with CR than those with PR only [19,27]. In contrast, others did not find any significant differences [9]. Baseline renal function, proteinuria, hypertension, histological activity and chronicity indexes have been previously suggested as predictors of renal response [9,18,19,24,29,30,31]. In our study, baseline proteinuria >1.5 g/day emerged as the most significant predictor of early (i.e., at 3, 6, 9 months) and late complete response (i.e., at 12, 18 and 24 months). Time to response has not been fully addressed in previous studies, with some reporting a correlation with improved renal survival [28] and others a predictive role of baseline proteinuria [18,32]. Our study predicted time to CR only by baseline proteinuria >1.5 g/day.
Renal flare is a recognized predictor of progressive CKD and morbidity in LN patients [10,33]. Different studies report flare rates of 25–66% [14,34,35,36], largely depending on follow-up time and definitions of flare. In our study, 33% of patients experienced a relapse in a median time of 38 months. Few data are available regarding factors predictive of flare. Attainment of CR, rather than PR, shorter time to CR and maintenance treatment with MPA (compared to AZA) have been the main determinants of flare occurrence across studies [14,35,36,37]. In our study, although a longer time to either CR or PR correlated with a higher risk of flare, this correlation was significant only for responses at 12, 18 and 24 months. Induction treatment with MPA (compared to CYC) and proteinuria <0.8 g/day at 12 months were significantly associated with a lower risk of flare, with the latter finding adding to the value of 12-month proteinuria as a predictor of renal outcome. Proteinuria at the time of LN diagnosis, apart from affecting time to response, was also found to be a risk factor for flare. The cut-off level that was strongly associated with a higher flare risk was determined at 2 g/day. Baseline proteinuria >2 g/day and 12-month proteinuria >0.8 g/day were also found to correlate with a shorter time to flare. Interestingly, pure proliferative classes conferred a greater risk of flare when compared to mixed classes, but the difference was significant only between class III and mixed classes. Other baseline histological characteristics did not affect the risk of flare. However, studies have shown that the activity index of repeat, instead of initial, biopsy may predict flare occurrence and the time of flare [38,39].
Recent studies report cumulative renal survival rates in proliferative LN of 91, 81, 75 and 66% at 5, 10, 15 and 20 years, respectively, and 5-, 10- and 15-year cumulative incidence of ESRD of 3–11%, 6–19% and 19–25% respectively [40,41,42,43]. In our study, 22% of patients reached the composite renal outcome (stage 3–4 CKD or ESRD) in a median follow-up time of 100 months. Several studies have identified baseline parameters as predictors of long-term renal prognosis, such as creatinine, proteinuria, hypertension, activity and chronicity index [43,44], while others [15,16,18,28,40,44,45] highlight the importance of renal parameters at 12 months as single variables (i.e., proteinuria <0.7 g/day or <0.8 g/day) or as composite endpoint (i.e., renal response). Renal response definition, however, is not uniform across studies. The current study demonstrated that the only variables strongly associated with long-term adverse renal outcomes were proteinuria >0.8 g/day at 12 months and interstitial fibrosis/tubular atrophy >25% at the initial biopsy. Baseline proteinuria did not seem to affect renal survival significantly. Some studies have reported that activity and chronicity index, as well as interstitial fibrosis/tubular atrophy, correlate with a worse renal outcome when evaluated in repeat and not in initial biopsies [38,46]. In our study, interstitial fibrosis/tubular atrophy emerged as the only histologic parameter of the initial biopsy that could predict a worse renal outcome. Interstitial fibrosis at initial biopsy has already been associated with adverse renal outcomes [45,47].These results imply that evaluating distinct components of the activity and chronicity indexes in initial and repeat biopsies may be of greater value in predicting renal response and long-term outcome than relying solely on activity and chronicity indexes. While some studies report that a very early response (i.e., at 3 months) can predict long-term renal outcomes [48,49], we did not find that such an early response significantly affected renal survival. In our study, a response at 6, 12, 18 and 24 months predicted a better renal outcome.
The use of an inception cohort of biopsy-proven proliferative (only) LN patients, the long follow-up time of 100 months, and the uniform approach in the setting of a specialized academic center constitute the main strengths of our study. The study’s limitations are its retrospective design and the inclusion of almost exclusively Caucasian patients.

5. Conclusions

The current study highlights the value of baseline proteinuria <1.5 g/day as the only early predictor of time to complete response in patients with proliferative LN. It also demonstrates that induction treatment with mycophenolic acid significantly lowers the risk of renal flare. At the same time, 12-month proteinuria >0.8 g/day correlates significantly with flare occurrence and, along with interstitial fibrosis/tubular atrophy >25% at the initial biopsy, strongly predicts long-term renal outcome.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm11175017/s1, Table S1: Differences in baseline characteristics between the two major induction treatment groups (mycophenolic acid versus cyclophosphamide); Table S2: Predictors of complete response at 3-6-9-12-18-24 months; Table S3: Response (CR or PR) at different time points as predictors of flare; Table S4: Statistical tests examining correlation between independent predictors of risk of flare; Table S5: Renal function at presentation, response to treatment and time to renal failure in patients with ESRD; Table S6: Responses at different time points as predictors of adverse renal outcome; Table S7: Statistical tests examining correlation between independent predictors of risk of adverse renal outcome; Figure S1: Kaplan-Meier survival estimates of probability for response according to [a] proliferative lupus nephritis class [b] treatment with cyclophosphamide versus MPA/MMF.

Author Contributions

Conceptualization, M.G.T.,J.B. and P.P.S.; Methodology, E.K., M.G.T. and I.M.; Software, I.M.; Validation, E.K., I.M., S.M., P.P.S., J.B. and M.G.T.; Formal Analysis, I.M.; Investigation, E.K., S.M. and M.G.T.; Resources, E.K., S.M., G.L., P.P.S., J.B. and M.G.T.; Data Curation, E.K. and I.M.; Writing—Original Draft Preparation, E.K. and M.G.T.; Writing—Review & Editing, S.M., G.L., I.M., P.P.S. and J.B.; Visualization, E.K.; Supervision, M.G.T. and S.M.; Project Administration, M.G.T. and J.B.; Funding Acquisition, none.All authors have read and agreed to the published version of the manuscript.

Funding

No funding was received for this study.

Institutional Review Board Statement

The study was approved by the Institutional Review Board (protocol number 1725/14-12-2017) of Laiko General Hospital of Athens. The analysis included anonymized study data, and no patient consent was required.

Informed Consent Statement

Patients and/or the public were not involved in the design, conduct, reporting or dissemination plans of this research.

Data Availability Statement

The data supporting this study’s findings are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Anders, H.-J.; Saxena, R.; Zhao, M.-H.; Parodis, I.; Salmon, J.E.; Mohan, C. Lupus nephritis. Nat. Rev. Dis. Primers 2020, 6, 7. [Google Scholar] [CrossRef] [PubMed]
  2. Doria, A.; Iaccarino, L.; Ghirardello, A.; Zampieri, S.; Arienti, S.; Sarzi-Puttini, P.; Atzeni, F.; Piccoli, A.; Todesco, S. Long-term prognosis and causes of death in systemic lupus erythematosus. Am. J. Med. 2006, 119, 700–706. [Google Scholar] [CrossRef] [PubMed]
  3. Yap, D.Y.; Tang, C.S.; Ma, M.K.; Lam, M.F.; Chan, T.M. Survival analysis and causes of mortality in patients with lupus nephritis. Nephrol. Dial. Transplant. 2012, 27, 3248–3254. [Google Scholar] [CrossRef] [PubMed]
  4. Lerang, K.; Gilboe, I.M.; Steinar Thelle, D.; Gran, J.T. Mortality and years of potential life loss in systemic lupus erythematosus: A population-based cohort study. Lupus 2014, 23, 1546–1552. [Google Scholar] [CrossRef] [PubMed]
  5. Costenbader, K.H.; Desai, A.; Alarcón, G.S.; Hiraki, L.T.; Shaykevich, T.; Brookhart, M.A.; Massarotti, E.; Lu, B.; Solomon, D.H.; Winkelmayer, W.C. Trends in the incidence, demographics, and outcomes of end-stage renal disease due to lupus nephritis in the US from 1995 to 2006. Arthritis Rheum. 2011, 63, 1681–1688. [Google Scholar] [CrossRef]
  6. Tektonidou, M.; Dasgupta, A.; Ward, M.M. Risk of end-stage renal disease in patients with lupus nephritis, 1971-2015: A systematic review and Bayesian meta-analysis. Arthritis Rheumatol. 2016, 68, 1432–1441. [Google Scholar] [CrossRef]
  7. Bastian, H.M.; Roseman, J.M.; McGwin, G.; Alarcón, G.S.; Friedman, A.W.; Fessler, B.J.; A Baethge, B.; Reveille, J.D.; LUMINA Study Group. Systemic lupus erythematosus in three ethnic groups. XII. Risk factors for lupus nephritis after diagnosis. Lupus 2002, 11, 152–160. [Google Scholar] [CrossRef]
  8. Bertsias, G.K.; Tektonidou, M.; Amoura, Z.; Aringer, M.; Bajema, I.; Berden, J.H.; Boletis, J.; Cervera, R.; Dörner, T.; Doria, A.; et al. Joint European League Against Rheumatism and European Renal Association–European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of adult and paediatric lupus nephritis. Ann. Rheum. Dis. 2012, 71, 1771–1782. [Google Scholar] [CrossRef]
  9. Almalki, A.H.; Alrowaie, F.A.; Alhozali, H.M.; Almalki, N.K.; Alsubei, A.I.; Alturki, M.S.; Sadagah, L.F. Remission and long-term outcomes of proliferative lupus nephritis: Retrospective study of 96 patients from Saudi Arabia. Lupus 2019, 28, 1082–1090. [Google Scholar] [CrossRef]
  10. Parikh, S.V.; Nagaraja, H.N.; Hebert, L.; Rovin, B.H. Renal flare as a predictor of incident and progressive CKD in patients with lupus nephritis. Clin. J. Am. Soc. Nephrol. 2014, 9, 279–284. [Google Scholar] [CrossRef]
  11. Gladman, D.D.; Ibañez, D.; Urowitz, M.B. Systemic Lupus Erythematosus Disease Activity Index 2000. J. Rheumatol. 2002, 29, 288–291. [Google Scholar] [PubMed]
  12. Kidney Disease: Improving Global Outcomes (KDIGO) Glomerulonephritis Work Group. KDIGO Clinical Practice Guideline for Glomerulonephritis. Kidney Int. Suppl. 2012, 2, 139–274. [Google Scholar]
  13. Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group. KDIGO clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int. Suppl. 2013, 3, 1–150. [Google Scholar]
  14. Ichinose, K.; Kitamura, M.; Sato, S.; Eguchi, M.; Okamoto, M.; Endo, Y.; Tsuji, S.; Takatani, A.; Shimizu, T.; Umeda, M.; et al. Complete renal response at 12 months after induction therapy is associated with renal relapse-free rate in lupus nephritis: Asinglecenter, retrospective cohort study. Lupus 2019, 28, 501–509. [Google Scholar] [CrossRef]
  15. Dall’Era, M.; Cisternas, M.G.; Smilek, D.E.; Straub, L.; Houssiau, F.A.; Cervera, R.; Rovin, B.H.; Mackay, M. Predictors of long-term renal outcome in lupus nephritis trials: Lessons learned from the Euro-Lupus nephritis cohort. Arthritis Rheumatol. 2015, 67, 1305–1313. [Google Scholar] [CrossRef] [PubMed]
  16. Tamirou, F.; Lauwerys, B.R.; Dall’Era, M.; Mackay, M.; Rovin, B.; Cervera, R.; AHoussiau, F. A proteinuria cut-off level of 0.7 g/day after 12 months of treatment best predicts long-term renal outcome in lupus nephritis: Data from the MAINTAIN Nephritis Trial. Lupus Sci. Med. 2015, 2, e000123. [Google Scholar] [CrossRef]
  17. Ugolini-Lopes, M.R.; Seguro, L.P.C.; Castro, M.X.F.; Daffre, D.; Lopes, A.C.; Borba, E.F.; Bonfá, E. Early proteinuria response: A valid real-life situation predictor of long-term lupus renal outcome in an ethnically diverse group with severe biopsy-proven nephritis? Lupus Sci. Med. 2017, 4, e000213. [Google Scholar] [CrossRef]
  18. Moroni, G.; Gatto, M.; Tamborini, F.; Quaglini, S.; Radice, F.; Saccon, F.; Frontini, G.; Alberici, F.; Sacchi, L.; Binda, V.; et al. Lack of EULAR/ERA-EDTA response at 1 year predicts poor long-term renal outcome in patients with lupus nephritis. Ann. Rheum. Dis. 2020, 79, 1077–1083. [Google Scholar] [CrossRef]
  19. Chen, Y.E.; Korbet, S.M.; Katz, R.S.; Schwartz, M.M.; Lewis, E.J. Value of a complete or partial remission in severe lupus nephritis. Clin. J. Am. Soc. Nephrol. 2008, 3, 46–53. [Google Scholar] [CrossRef]
  20. Ginzler, E.M.; Wofsy, D.; Isenberg, D.; Gordon, C.; Lisk, L.; Dooley, M.A. Mycophenolate mofetil or intravenous cyclophosphamide for induction treatment for lupus nephritis. N. Engl. J. Med. 2005, 353, 2219–2228. [Google Scholar] [CrossRef]
  21. Appel, G.B.; Contreras, G.; Dooley, M.A.; Ginzler, E.M.; Isenberg, D.; Jayne, D.; Li, L.-S.; Mysler, E.; Sánchez-Guerrero, J.; Solomons, N.; et al. Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J. Am. Soc. Nephrol. 2009, 20, 1103–1112. [Google Scholar] [CrossRef] [PubMed]
  22. Korbet, S.M.; Lewis, E.J.; Collaborative Study Group. Complete remission in severe lupus nephritis: Assessing the rate of loss in proteinuria. Nephrol. Dial. Transplant. 2012, 27, 2813–2819. [Google Scholar] [CrossRef] [Green Version]
  23. Liu, G.; Wang, H.; Le, J.; Lan, L.; Xu, Y.; Yang, Y.; Chen, J.; Han, F. Early-stage predictors for treatment responses in patients with active lupus nephritis. Lupus 2019, 28, 283–289. [Google Scholar] [CrossRef] [PubMed]
  24. Luís, M.S.; Bultink, I.E.; da Silva, J.A.; Voskuyl, A.E.; Inês, L.S. Early predictors of renal outcome in patients with proliferative lupus nephritis:a 36-month cohort study. Rheumatology 2021, 60, 5134–5141. [Google Scholar] [CrossRef] [PubMed]
  25. Fernandes das Neves, M.; Irlapati RV, P.; Isenberg, D. Assessment of long-term remission in lupus nephritis patients: A retrospective analysis over 30 years. Rheumatology 2015, 54, 1403–1407. [Google Scholar] [CrossRef]
  26. Pakchotanon, R.; Gladman, D.D.; Su, J.; Urowitz, M.B. Sustained complete renal remission is a predictor of reduced mortality, chronic kidney disease and endstage renal disease in lupus nephritis. Lupus 2018, 27, 468–474. [Google Scholar] [CrossRef]
  27. Koo, H.S.; Kim, S.; Chin, H.J. Remission of proteinuria indicates good prognosis in patients with diffuse proliferative lupus nephritis. Lupus 2016, 25, 3–11. [Google Scholar] [CrossRef]
  28. Pirson, V.; Enfrein, A.; Houssiau, F.A.; Tamirou, F. Absence of renal remission portends poor long-term kidney outcome in lupus nephritis. Lupus Sci. Med. 2021, 8, e000533. [Google Scholar] [CrossRef]
  29. Moroni, G.; Quaglini, S.; Radice, A.; Trezzi, B.; Raffiotta, F.; Messa, P.; Sinico, R.A. The value of a panel of autoantibodies for predicting the activity of lupus nephritis at time of renal biopsy. J. Immunol. Res. 2015, 2015, 106904. [Google Scholar] [CrossRef]
  30. Ichinose, K.; Kitamura, M.; Sato, S.; Fujikawa, K.; Horai, Y.; Matsuoka, N.; Tsuboi, M.; Nonaka, F.; Shimizu, T.; Fukui, S.; et al. Factors predictive of long-term mortality in lupus nephritis: A multicenter retrospective study of a Japanese cohort. Lupus 2019, 28, 295–303. [Google Scholar] [CrossRef]
  31. McDonald, S.; Yiu, S.; Su, L.; Gordon, C.; Truman, M.; Lisk, L.; Solomons, N.; Bruce, I.N. Predictors of treatment response in a lupus nephritis population: Lessons from the Aspreva Lupus Management Study (ALMS) trial. Lupus Sci. Med. 2022, 9, e000584. [Google Scholar] [CrossRef] [PubMed]
  32. Touma, Z.; Urowitz, M.B.; Ibañez, M.; Gladman, D.D. Time to recovery from proteinuria in patients with lupus nephritis receiving standard treatment. J. Rheumatol. 2014, 41, 688–697. [Google Scholar] [CrossRef] [Green Version]
  33. Mosca, M.; Bencivelli, W.; Neri, R.; Pasquariello, A.; Batini, V.; Puccini, R.; Tavoni, A.; Bombardieri, S. Renal flares in 91 SLE patients with diffuse proliferative glomerulonephritis. Kidney Int. 2002, 61, 1502–1509. [Google Scholar] [CrossRef] [PubMed]
  34. Sprangers, B.; Monahan, M.; Appel, G.B. Diagnosis and treatment of lupus nephritis flares—an update. Nat. Rev. Nephrol. 2012, 8, 709–717. [Google Scholar] [CrossRef] [PubMed]
  35. Illei, G.G.; Takada, K.; Parkin, D.; Austin, H.A.; Crane, M.; Yarboro, C.H.; Vaughan, E.M.; Kuroiwa, T.; Danning, C.L.; Pando, J.; et al. Renal flares are common in patients with severe proliferative lupus nephritis treated with pulse immunosuppressive therapy: Long-term followup of a cohort of 145 patients participating in randomized controlled studies. Arthritis Rheum. 2002, 46, 995–1002. [Google Scholar] [CrossRef]
  36. Yap, D.Y.; Tang, C.; Ma, M.K.; Mok, M.M.; Chan, G.C.; Kwan, L.P.; Chan, T.M. Longterm data on disease flares in patients with proliferative lupus nephritis in recent years. J. Rheumatol. 2017, 44, 1375–1383. [Google Scholar] [CrossRef] [PubMed]
  37. So, M.W.; Koo, B.S.; Kim, Y.G.; Lee, C.-K.; Yoo, B. Predictive value of remission status after 6 months induction therapy in patients with proliferative lupus nephritis: A retrospective analysis. Clin. Rheumatol. 2011, 30, 1399–1405. [Google Scholar] [CrossRef] [PubMed]
  38. Parodis, I.; Adamichou, C.; Aydin, S.; Gomez, A.; Demoulin, N.; Weinmann-Menke, J.; A Houssiau, F.; Tamirou, F. Per-protocol repeat kidney biopsy portends relapse and long-term outcome in incident cases of proliferative lupus nephritis. Rheumatology 2020, 59, 3424–3434. [Google Scholar] [CrossRef] [PubMed]
  39. De Rosa, M.; Azzato, F.; Toblli, J.E.; De Rosa, G.; Fuentes, F.; Nagaraja, H.N.; Nash, R.; Rovin, B.H. A prospective observational cohort study highlights kidney biopsy findings of lupus nephritis patients in remission who flare following withdrawal of maintenance therapy. Kidney Int. 2018, 94, 788–794. [Google Scholar] [CrossRef]
  40. Farinha, F.; Pepper, R.J.; Oliveira, D.G.; McDonnell, T.; Isenberg, D.A.; Rahman, A. Outcomes of membranous and proliferative lupus nephritis–analysis of a single-centre cohort with more than 30 years of follow-up. Rheumatology 2020, 59, 3314–3323. [Google Scholar] [CrossRef]
  41. Mahajan, A.; Amelio, J.; Gairy, K.; Kaur, G.; A Levy, R.; Roth, D.; Bass, D. Systemic lupus erythematosus, lupus nephritis and end-stage renal disease: A pragmatic review mapping disease severity and progression. Lupus 2020, 29, 1011–1020. [Google Scholar] [CrossRef] [PubMed]
  42. Hanly, J.G.; O’Keeffe, A.G.; Su, L.; Urowitz, M.B.; Romero-Diaz, J.; Gordon, C.; Bae, S.-C.; Bernatsky, S.; Clarke, A.E.; Wallace, D.J.; et al. The frequency and outcome of lupus nephritis: Results from an international inception cohort study. Rheumatology 2016, 55, 252–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Moroni, G.; Vercelloni, P.G.; Quaglini, S.; Gatto, M.; Gianfreda, D.; Sacchi, L.; Raffiotta, F.; Zen, M.; Costantini, G.; Urban, M.L.; et al. Changing patterns in clinical–histological presentation and renal outcome over the last five decades in a cohort of 499 patients with lupusnephritis. Ann. Rheum. Dis. 2018, 77, 1318–1325. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, J.; Liang, D.; Zhang, H.; Liu, Z.; Le, W.; Zhou, M.; Hu, W.; Zeng, C. Long-term renal outcomes in a cohort of 1814 Chinese patients with biopsy-proven lupus nephritis. Lupus 2015, 24, 1468–1478. [Google Scholar] [CrossRef]
  45. Vajgel, G.; Oliveira, C.B.L.; Costa, D.M.N.; Cavalcante, M.A.G.M.; Valente, L.M.; Sesso, R.; Crovella, S.; Kirsztajn, G.M.; Sandrin-Garcia, P. Initial renal histology and early response predict outcomes of Brazilian lupus nephritis patients. Lupus 2020, 29, 83–91. [Google Scholar] [CrossRef]
  46. Gatto, M.; Radice, F.; Saccon, F.; Calatroni, M.; Frontini, G.; Trezzi, B.; Zen, M.; Ghirardello, A.; Tamborini, F.; Binda, V.; et al. Clinical and histological findings at second but not at first kidney biopsy predict end-stage kidney disease in a large multicentric cohort of patients with active lupus nephritis. Lupus Sci. Med. 2022, 9, 000689. [Google Scholar] [CrossRef]
  47. Wilson, P.C.; Kashgarian, M.; Moeckel, G. Interstitial inflammation and interstitial fibrosis and tubular atrophy predict renal survival in lupus nephritis. Clin. Kidney J. 2018, 11, 207–218. [Google Scholar] [CrossRef]
  48. Tamirou, F.; D’Cruz, D.; Sangle, S.; Remy, P.; Vasconcelos, C.; Fiehn, C.; Guttierez, M.D.M.A.; Gilboe, I.-M.; Tektonidou, M.; Blockmans, D.; et al. Long-term follow-up of the MAINTAIN Nephritis Trial, comparing azathioprine and mycophenolate mofetil as maintenance therapy of lupus nephritis. Ann. Rheum. Dis. 2016, 75, 526–531. [Google Scholar] [CrossRef]
  49. Hanaoka, H.; Yamada, H.; Kiyokawa, T.; Iida, H.; Suzuki, T.; Yamasaki, Y.; Ooka, S.; Nagafuchi, H.; Okazaki, T.; Ichikawa, D.; et al. Lack of partial renal response by 12 weeks after induction therapy predicts poor renal response and systemic damage accrual in lupus nephritis class III or IV. Arthritis Res. Ther. 2017, 19, 4. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Patients with a renal response at different time points during follow-up.
Figure 1. Patients with a renal response at different time points during follow-up.
Jcm 11 05017 g001
Figure 2. Kaplan–Meier survival estimates of probability for flare according to (a) Proteinuria at the time of LN diagnosis, (b) Proteinuria at 12 months.
Figure 2. Kaplan–Meier survival estimates of probability for flare according to (a) Proteinuria at the time of LN diagnosis, (b) Proteinuria at 12 months.
Jcm 11 05017 g002
Table 1. Baseline demographic, clinical, laboratory and histological characteristics and immunosuppressive treatment regimens.
Table 1. Baseline demographic, clinical, laboratory and histological characteristics and immunosuppressive treatment regimens.
Baseline CharacteristicsMean ± SD,
Median(IQR),
N/%
Age (yr) mean ± SD31 ± 13
Sex (M-F) N/%20/20–80/80
Race (Caucasian-Other) N/%96/96–4/4
Time from SLE diagnosis to LN (years)median(IQR)()
LN as first presentation of SLE N/%51/51
SLEDAI score median(IQR)12(4)
Low C3 N/% 165/77.5
Low C4 N/% 155/65.5
Positive anti-dsDNA antibodies N/% 263/78.5
Proteinuria (g/24 h) median (IQR)2.6(4)
  • Proteinuria >3 g/d N/%
48/48
  • Proteinuria 1–3 g/d N/%
31/31
  • Proteinuria <1 g/d N/%
21/21
Active urine sediment N/%92/92
Hypertension N/%28/28
Serum albumin (g/dL) mean ± SD3.1 ± 0.8
Serum Cr (mg/dL) median(IQR)0.8(0.5)
eGFR (mL/min/1.73 m2) median(IQR)94.5(50)
  • eGFR >60 N/%
75/75
  • eGFR 30–60 N/%
14/14
  • eGFR <30 N/%
11/11
LN class
  • III N/%
28/28
  • IV N/%
47/47
  • III + V N/%
9/9
  • IV + V N/%
16/16
Number of crescents median(IQR)2(4)
Activity Index median(IQR)10(6)
Chronicity Index median(IQR)2(2)
Interstitial fibrosis/tubular atrophy *,3
  • <25% N/%
85/87
  • >25% N/%
13/13
Induction Treatment
  • mycophenolic acid N/%
27/27
  • cyclophosphamide N/%
69/69
  • other
3/3
  • none N/%
1/1
Maintenance Treatment
  • mycophenolic acid N/%
77/77
  • azathioprine N/%
8/8
  • cyclophosphamide N/%
5/5
  • none N/%
10/10
Duration of total treatment (months)median(IQR)39(38)
eGFR: estimated glomerular filtration rate using the CKD-EPI formula, SLEDAI: systemic lupus erythematosus disease activity index, anti-ds DNA: antibodies against double-stranded DNA. * refers to the percentage of the renal cortex involved by interstitial fibrosis and tubular atrophy. 1 data available for 84/100 patients, 2 data available for 80/100 patients, 3 data available for 98/100 patients.
Table 2. Predictors of time to complete response.
Table 2. Predictors of time to complete response.
VariablesUnivariate ModelsMultivariate Model
HR95% Cis
(p-Value)
HR95% Cis
(p-Value)
Age (years)1.010.98, 1.02 (0.53)
Sex
  • Male
Reference Group
  • Female
1.290.73, 2.29 (0.36)
Time from SLE diagnosis to LN (years)0.980.94, 1.03 (0.6)
Hypertension
  • No
Reference Group
  • Yes
0.70.43, 1.16 (0.18)
Low C3
  • Yes
Reference Group
  • No
0.730.40, 1.33 (0.31)
Low C4
  • Yes
Reference Group
  • No
0.680.4, 1.15 (0.15)
Anti-dsDNA antibodies
  • Negative
Reference Group
  • Positive
1.260.7, 2.32 (0.44)
eGFR at diagnosis(mL/min/1.73 m2)
  • >60
Reference Group
  • <60
0.660.39, 1.13 (0.13)
Proteinuria at diagnosis (g/day)
  • >1.5
Reference Group
  • <1.5
1.771.10, 2.84 (0.01)
LN class
  • III
Reference Group
  • IV
0.870.5, 1.47 (0.62)
  • III/IV + V
0.690.37, 1.31 (0.26)
Number of Crescents0.970.93, 1.01 (0.24)
Activity Index10.96, 1.05 (0.75)
Chronicity Index0.920.82, 1.04 (0.19)
Interstitial fibrosis/Tubular atrophy
  • <25% *
Reference Group
  • >25% *
0.820.39, 1.72 (0.61)
Induction treatment **
  • CYC
Reference Group
  • MPA
0.920.55, 1.56 (0.78)
eGFR: estimated glomerular filtration rate using the CKD-EPI formula, SLEDAI: systemic lupus erythematosus disease activity index, anti-ds DNA: antibodies against double-stranded DNA, CYC: cyclophosphamide, MPA: mycophenolic acid. * refers to the percentage of the renal cortex involved by interstitial fibrosis and tubular atrophy, ** the two treatment groups were compared with and without the concomitant use of rituximab.
Table 3. Predictors of flares.
Table 3. Predictors of flares.
VariablesUnivariate ModelsMultivariate Model
OR95% CIs
(p-Value)
OR95% CIs
(p-Value)
Age (years)0.950.92, 0.98 (0.02)0.980.94, 1.03 (0.58)
Sex
  • Male
Reference Group
  • Female
0.50.19, 1.42 (0.2)
Time from SLE diagnosis to LN (years)0.930.84, 1.04 (0.23)
Hypertension
  • No
Reference Group
  • Yes
0.960.37, 2.47 (0.94)
Low C3
  • Yes
Reference Group
  • No
0.90.29, 2.96 (0.9)
Low C4
  • Yes
Reference Group
  • No
0.30.09, 0.99 (0.05)
Anti-dsDNA antibodies
  • Negative
Reference Group
  • Positive
0.960.29, 3.1 (0.94)
eGFR at diagnosis (mL/min/1.73 m2)
  • >60
Reference Group
  • <60
0.70.27, 1.98 (0.5)
Proteinuria at diagnosis (g/day)
  • <2
Reference Group
  • >2
31.14, 7.89 (0.02)
LN class
  • III
Reference Group
  • IV
0.590.22, 1.55 (0.28)0.380.09, 1.6 (0.19)
  • III/IV + V
0.210.05, 0.8 (0.02)0.130.01, 0.8 (0.02)
Number of Crescents0.960.88, 1.05 (0.44)
Activity Index1.010.91, 1.11 (0.8)
Chronicity Index0.770.59, 1.01 (0.06)
Interstitial fibrosis/Tubular atrophy
  • <25% *
Reference Group
  • >25% *
0.60.15, 2.4 (0.48)
Induction treatment **
  • CYC
Reference Group
  • MPA
0.250.08, 0.8 (0.02)0.140.03, 0.7 (0.01)
Time to PR (months)1.141.02, 1.26 (0.01)
Time to CR (months)1.051.01, 1.1 (0.01)
Time to CR/PR (months)1.111.02, 1.21 (0.01)
Proteinuria at 12 m (g/day)
  • <0.8
Reference Group
  • >0.8
3.381.14, 10 (0.02)4.121.15, 14 (0.02)
eGFR: estimated glomerular filtration rate using the CKD-EPI formula, SLEDAI: systemic lupus erythematosus disease activity index, anti-ds DNA: antibodies against double-stranded DNA, CYC: cyclophosphamide, MPA: mycophenolic acid, CR: complete response, PR: partial response. * refers to the percentage of the renal cortex involved by interstitial fibrosis and tubular atrophy, ** the two treatment groups were compared with and without the concomitant use of rituximab.
Table 4. Predictors of long-term adverse renal outcome (stage 3–4 CKD, ESRD).
Table 4. Predictors of long-term adverse renal outcome (stage 3–4 CKD, ESRD).
VariablesUnivariate ModelsMultivariate Model
OR95% CIs
(p-Value)
OR95% CIs
(p-Value)
Age (years)10.96, 1.03 (0.9)
Sex
  • Male
Reference Group
  • Female
0.80.2, 2.5 (0.7)
Time from SLE diagnosis to LN (years)10.9, 1.1 (0.9)
Hypertension
  • No
Reference Group
  • Yes
2.280.83, 6.2 (0.1)
Low C3
  • Yes
Reference Group
  • No
0.750.2, 3 (0.68)
Low C4
  • Yes
Reference Group
  • No
1.60.53, 4.9 (0.4)
Anti-dsDNA antibodies
  • Negative
Reference Group
  • Positive
0.450.1, 1.5 (0.2)
eGFR at diagnosis (mL/min/1.73 m2)
  • >60
Reference Group
  • <60
62, 16 (0.001)
Proteinuria at diagnosis (g/day)
  • <1.5
Reference Group
  • >1.5
1.70.56, 5.12 (0.34)
LN class
  • III
Reference Group
  • IV
0.590.19, 1.77 (0.34)
  • III/IV + V
0.620.17, 2.24 (0.47)
Number of Crescents10.93, 1.07 (0.91)
Activity Index1.030.92, 1.16 (0.5)
Chronicity Index1.130.87, 1.47 (0.34)
Interstitial fibrosis/Tubular atrophy
  • <25% *
Reference Group
  • >25% *
5.441.59, 18 (0.007)7.71.48, 40 (0.01)
Induction treatment **
  • CYC
Reference Group
  • MPA
1.020.35, 3 (0.95)
Time to PR (months)1.141.04, 1.26 (0.006)
Time to CR (months)1.040.99, 1.1 (0.1)
Time to CR/PR (months)1.11.02, 1.2 (0.01)
Proteinuria at 12 m (g/day)
  • <0.8
Reference Group
  • >0.8
9.51.84, 20 (0.003)10.82.7, 42 (0.001)
eGFR: estimated glomerular filtration rate using the CKD-EPI formula, SLEDAI: systemic lupus erythematosus disease activity index, anti-ds DNA: antibodies against double-stranded DNA, CYC: cyclophosphamide, MPA: mycophenolic acid, CR: complete response, PR: partial response. * refers to the percentage of the renal cortex involved by interstitial fibrosis and tubular atrophy, ** the two treatment groups were compared with and without the concomitant use of rituximab.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kapsia, E.; Marinaki, S.; Michelakis, I.; Liapis, G.; Sfikakis, P.P.; Boletis, J.; Tektonidou, M.G. Predictors of Early Response, Flares, and Long-Term Adverse Renal Outcomes in Proliferative Lupus Nephritis: A 100-Month Median Follow-Up of an Inception Cohort. J. Clin. Med. 2022, 11, 5017. https://doi.org/10.3390/jcm11175017

AMA Style

Kapsia E, Marinaki S, Michelakis I, Liapis G, Sfikakis PP, Boletis J, Tektonidou MG. Predictors of Early Response, Flares, and Long-Term Adverse Renal Outcomes in Proliferative Lupus Nephritis: A 100-Month Median Follow-Up of an Inception Cohort. Journal of Clinical Medicine. 2022; 11(17):5017. https://doi.org/10.3390/jcm11175017

Chicago/Turabian Style

Kapsia, Eleni, Smaragdi Marinaki, Ioannis Michelakis, George Liapis, Petros P. Sfikakis, John Boletis, and Maria G. Tektonidou. 2022. "Predictors of Early Response, Flares, and Long-Term Adverse Renal Outcomes in Proliferative Lupus Nephritis: A 100-Month Median Follow-Up of an Inception Cohort" Journal of Clinical Medicine 11, no. 17: 5017. https://doi.org/10.3390/jcm11175017

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop