Abstract
Purpose of review
Despite recent developments and treatment successes, the outcome and prognosis of patients with lupus nephritis have not greatly changed since the 1980s. This review covers the application of new concepts to the understanding of renal inflammation and the study of new pharmacologic agents to improve patient outcomes.
Recent findings
Studies have shown that the presence of anti-vimentin antibodies and T follicular helper cells in patient biopsies are associated with more severe interstitial inflammation, which has been tied to faster disease progression and onset of end stage renal disease. Additionally, data regarding the role of serum IgE anti-dsDNA antibodies in lupus nephritis by means of mediating type I inferferon production by plasmacytoid dendritic cells is highlighted. Finally, a thorough review of completed and currently open clinical trials of therapeutic agents is provided.
Summary
Current management of lupus nephritis is guided almost exclusively by glomerular involvement. Based on the data provided in this review, we argue that renal tubulointerstitial inflammation is no less important and represents an overlooked feature in the current clinical approach to patients. Tubulointerstitial inflammation is driven by both adaptive and innate immune mechanisms that are still poorly understood. Studying these pathogenic processes promises to reveal new therapeutic opportunities for those lupus nephritis patients with the worse prognosis.
Keywords: lupus nephritis, tubulointerstitial inflammation, glomerulonephritis
Introduction
Lupus nephritis (LuN) is the most common severe manifestation of systemic lupus erythematosus (SLE)(1, 2) and contributes significantly to mortality in this disease(3–5). Up to 50% of SLE patients develop LuN, which usually requires potentially toxic immunosuppressive therapies(6–8). Despite currently available aggressive treatments, up to 50% of patients progress to end stage renal disease (ESRD) within 5 years of diagnosis(9–11). This response rate has not significantly improved since the 1980s, despite the advent of new therapies(8, 12, 13). This review focuses on available therapies as well as those undergoing current clinical trial investigation. In addition, an overview of the inflammatory responses, characterizing human LuN, is provided.
Current Treatment Paradigms
Current diagnostic criteria and clinical management of LuN, as recommended by the American College of Rheumatology (ACR) and European Union League Against Rheumatism (EULAR), almost exclusively focus on glomerulonephritis(14, 15). Decision-making for the initiation of treatment, selection of a therapy regimen, and duration of treatment are dependent on the characterization of renal involvement based on the 2003 International Society of Nephrology/Renal Pathology Society (ISN/RPS) classification criteria (Table 1)(16).
Table 1.
2003 ISN/RPS lupus nephritis classification, adapted from (16)
| Class | Type |
|---|---|
| I | Minimal mesangial LuN |
| II | Mesangial proliferative LuN |
| III | Focal LuN |
| IV | Diffuse segmental or global LuN |
| V | Membranous LuN |
| VI | Advanced sclerosing LuN |
In clinical practice, human lupus glomerulonephritis (GN) is treated with a regimen of induction therapy followed by additional maintenance immunosuppression. Many of the treatments under discussion in this review have not received either US or European regulatory agency approvals for the indication of lupus nephritis therapy and their use should be considered as off-label or investigational (FDA and EMA, respectively). Traditional regimens are based on the National Institutes of Health (NIH) trials, which found that glucocorticoids (GC) plus intravenous (IV) pulse cyclophosphamide (CYC) were more effective than glucocorticoids alone in LuN (all classes) (12). There is also data supporting low dose pulse IV CYC based on the Euro-Lupus (EL CYC) regimen where patients receive IV CYC 500 mg every 2 weeks for 6 doses (17, 18). Mycophenolate mofetil (MMF) is another recognized option for induction therapy. Multiple trials have demonstrated that MMF has comparable efficacy to CYC (8, 19). Additionally, in Black/mixed race patients, MMF showed higher response rates compared to IV cyclophosphamide (60.4 % vs 38.5%, p =0.033)(8).
After completion of initial induction therapy, maintenance therapy is generally indicated in all patients. Studies have shown that patients who are not placed on maintenance immunosuppressive therapy are at increased risk of worsening renal function, ESRD, and even death (20, 21). The ALMS study found treatment failure, renal flares, and time to rescue therapy were all significantly higher utilizing azathioprine (AZA) as compared to MMF for this indication (22). Conversely, the MAINTAIN trial (LuN class III/IV/V) found similar rates of renal flares (23) and these findings were maintained at 10 year follow-up (24). The optimal duration maintenance therapy with either agent is not clear at present. The ALE06 trial (Clinicaltrials.gov identifier: NCT01946880) is a multi-center randomized controlled trial (RCT), investigating the effects of MMF withdrawal in clinically stable SLE patients that is presently ongoing to help answer this important clinical question. Uncomplicated class V LuN is currently treated as a special subset with different treatment recommendations based on published trials (25, 26). The ACR and EULAR both currently recommend prednisone and MMF for induction in patients with isolated class V involvement (14, 15).
As noted in the above discussion, despite current therapies, a significant proportion of patients with LuN develop ESRD. Therefore, there is an unmet need for novel therapies (11, 27–29). Tacrolimus has been studied as a potential alternative for induction therapy. Chen et al. examined 81 patients with class III/IV/V LuN who were randomly assigned to tacrolimus vs NIH IV CYC regimen plus GC(30). While proteinuria was significantly reduced in the tacrolimus group, complete response (CR) rates were comparable (52.4% vs 38.5%, p=0.2). A more recent study by Mok et al compared GCs combined with either tacrolimus or MMF (n=150) followed by AZA in Chinese patients with class III/IV/V LuN (31), and found tacrolimus to be noninferior at 6 months. Studies have also looked at combining tacrolimus with MMF. Liu et al. compared 368 Chinese patients with class III/IV/V LuN treated with GCs and either tacrolimus and MMF or IV CYC every 4 weeks for 6 doses (32). At 6 months, 45.9% in the tacrolimus/MMF group had attained CR vs 25.6% in the IV CYC group (p<0.001) with similar percentages of adverse events (50.3 vs 52.5%). A 2016 meta-analysis by Zhang et al. found that calcineurin inhibitors (either tacrolimus or cyclosporine) were as effective as IV CYC (5 studies) or MMF (2 studies) for LuN (class III/IV/V) (33). There is a present ongoing, multicenter, RCT investigating the efficacy of tacrolimus vs MMF for induction therapy in LuN (Clinicaltrials.gov identifier: NCT01580865)
Given their success in other rheumatic diseases, biologic therapies have also been studied as potential treatments for lupus nephritis. Rituximab (RTX), which targets CD20 expressed on B cells, has been shown to be beneficial in multiple small open label and retrospective studies (34–39). Despite these encouraging results, LUNAR - a phase III RCT - produced contradictory results (40). This trial included 144 patients with class III/IV +/− V LuN treated with either RTX or placebo plus MMF and GCs. At 52 weeks there was no difference in CR or partial response (PR) rates in the RTX vs placebo groups (56.9% vs 45.8%, p=0.18). In a subgroup analysis, Black patients in the RTX group were found to have a trend to higher overall response rates (OR: combined PR and CR) compared to placebo (70% vs 45%, p=0.20).
Despite LUNAR, studies have continued to investigate the potential use of RTX in LuN. Moroni et al. performed a clinical observational study of 54 patients with class III/IV/V LuN induced with either RTX, MMF, or CYC (41). At 3 months, renal response rates were similar among the three groups (58.8% RTX vs 63.1% CYC vs 64.7% MMF). At 12 months, CR rates were also similar between the groups (70.6% RTX vs 65% CYC vs 52.9% MMF). Although this was a small, non-controlled trial, it suggests that RTX may still be beneficial in select LuN patients.
Belimumab, which inhibits B-cell activating factor (BAFF), is an FDA-approved agent for treating non-renal SLE that is also being studied as a potential therapy for LuN. The BLISS-52 and BLISS-76 trials focused on stable, non-severe LuN and excluded patients with acute and/or severe LuN defined as proteinuria ≥ 6g/day, serum creatinine > 2.5, or those on dialysis (42, 43). In a pooled analysis Dooley et al. found that although patients receiving belimumab 1 mg/kg or 10 mg/kg had decreased proteinuria compared to placebo, this difference did not meet clinical significance (median decrease: 48.3%/39.1% vs 75.2%, NS) (44). Likewise, there was no significant difference in renal flare (2.1%/1.1% vs 3%, NS) and renal remission rates (65.7%/70.5% vs 58.7%, NS) as compared to placebo. Despite this, renal improvement as measured by SELENA-SLEDAI(45), was statistically significant in patients being treated concurrently with MMF and belimumab 1 mg/kg and 10 mg/kg groups compared to placebo (52.6%/63.2% vs 27.8%, p=0.03). Although these studies only included patients with stable, non-severe LuN, they suggest that belimumab may be beneficial. There is a current phase III, double-blinded, RCT investigating the effects/safety of standard of care plus belimumab 10 mg/kg vs placebo in patients with LuN to address this question. (Clinicaltrials.gov identifier: NCT01639339) Another candidate biologic therapy for LuN is abatacept which is a soluble fusion protein of cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and Fc immunoblobulin IgG1. ACCESS randomized patients with class III/IV+/−V LuN to weight-based IV abatacept vs placebo (46). Both groups also received GCs and 500 mg IV CYC every 2 weeks for 6 doses, followed by AZA maintenance. At 24 weeks, there was no difference in CR or OR rates in the abatacept vs placebo groups (33% vs 31% CR, 59% vs 59% OR). Adverse events were comparable in both groups. Studies have also looked at combination abatacept and MMF. A double-blind study randomized patients with Class III/IV +/− V LuN to IV abatacept 30 mg/kg through day 57 then 10 mg/kg vs 10 mg/kg vs placebo (47). Patients were also maintained on MMF and GCs. Investigators found no difference between placebo and abatacept in regards to patients who met criteria for CR or PR. There is a current ongoing phase III, double-blind multicenter RCT investigating abatacept vs placebo with background MMF and GCs over a 104-week period. The primary efficacy endpoint is the proportion of patients attaining CR based on renal function, proteinuria, urine sediment, and GC dose (Clinicaltrials.gov identifier: NCT01714817).
There have been several other recent studies evaluating novel treatments for proliferative LuN, which were terminated early due to side effects or inefficacy. Atacicept is a soluble, fully human, recombinant fusion protein, which inhibits both BAFF and a proliferation-inducing ligand (APRIL)(48). A phase II/III double-blind RCT of subcutaneous atacicept (4 patients) or placebo (2 patients) plus MMF and prednisone for active class III/IV LuN was discontinued early due to severe, rapid declines in serum IgG and serious infections in 3 out 4 treated patients(49). Ocrelizumab, a recombinant, humanized antibody, sharing targeting of CD20 with rituximab (50), was also studied in a double- blind phase III RCT of 381 patients with class III/IV+/−V LuN (51). This study was also terminated early due to increased risk of serious infections (28.3% overall) in the ocrelizumab group. The long-term extension trial of BIIB023, a humanized monoclonal antibody against TNF-related weak induced of apoptosis (TWEAK), in class III/IV+/−V disease was also recently terminated due to inefficacy (Clinicaltrials.gov identifier: NCT01930890).
Of the possible new therapies undergoing current development, blisibimod is a BAFF antagonist that binds to both soluble and membrane-bound BAFF and inhibits the interaction of BAFF with BAFF receptor, transmembrane activator and CAML interactor (TACI), and B-cell maturation antigen (BCMA)(52). A recent double-blind phase Ia/Ib RCT of 56/64 patients with mild/inactive lupus were randomized to receive either blisibimod SC/IV, respectively, at various doses vs placebo (53). The trial found a significant decrease in patient serum B cell counts with the study drug without an increase in adverse events. While this trial excluded patients with active LuN, there is a pending phase III, double-blind RCT to evaluate the efficacy and safety of blisibimod in patients with lupus and stable LuN (Clinicaltrials.gov identifier: NCT02514967).
Mizoribine (MIZ) is another potential novel treatment for LuN. It is thought to inhibit inosine monophosphate dehydrogenase – the same target as MMF (54). Feng et al. randomized 90 Chinese patients with active Class III/IV/V LuN to receive MIZ every other day vs MMF vs IV CYC 500mg every 2 weeks plus GC over a 24-week period (55). All three groups showed an improvement in renal responses at 12 weeks but the CYC group had more early responders compared to MIZ (96.7% vs 73.3%, p=0.02). At 24 weeks, CR and OR rates were similar between all groups (CR- 22.7% MIZ, 24% MMF, 25% CYC, overall response - 68.2%, 72%, 75%). An ongoing phase III non-inferiority RCT is actively recruiting patients in China to further investigate the role of MIZ vs CYC in class III/IV/V LuN (Clinicaltrials.gov identifier: NCT02256150).
Type I inferferons, including interferon α, have been implicated in the pathogenesis of SLE. Anifrolumab, a human monoclonal antibody against the interferon α receptor 1(56), is currently being studied as a potential new therapy for lupus. A phase II open label trial in 17 Japanese patients with SLE showed that anifrolumab suppressed interferon gene signatures in 85% of patients at the 300 mg dose at day 169 and 95% of patients in the 1000 mg dose at day 85 without any safety signals(57). The TULIP-LN1 trial is a phase II double-blind RCT, which is currently enrolling patients to study the efficacy and safety of anifrolumab vs placebo in class III/IV +/− V LuN (Clinicaltrials.gov identifier: NCT02547922).
Finally, laquinimod (LAQ) is another promising novel therapy for LuN. Laquinimod’s method of action is not fully known but it is thought to behave as an oral immunomodulatory agent by downregulating select cytokines, including IL-6, IL-12, IL-17, IL-23, and TNFα while increasing IL-10(58). In a phase IIa double-blind RCT of 46 LuN patients comparing LAQ 0.5 mg or 1 mg daily vs placebo plus MMF and GCs(58), the investigators found that renal responses were higher in the LAQ 1 mg compared to the 0.5 mg and placebo groups (62.5% vs 40% vs 33.3%) at 24 weeks. The authors note they are planning a larger study to further investigate the efficacy and safety of LAQ but no formal trials have been registered at this time.
Pathophysiology and current experimental models
As previously noted, although kidney involvement in LuN can affect either the glomerular or tubulointerstitial compartments as well as combinations thereof, most classification schemes utilized in clinical practice focus almost exclusively on glomerular pathology. Two other commonly used tools for assessing disease severity – the NIH activity and chronicity indices (59) – are likewise heavily weighted towards glomerular, as opposed to tubulointerstitial involvement (Table 2) Moreover, fibrosis and sclerosis generally contribute more to the above scoring system than inflammation in either glomerular or tubulointerstitial compartments (Table 2)(60). A growing body of published literature supports the importance of tubulointerstitial inflammation (TII) in determining prognosis and patient outcomes(61–65). Although these studies noted an association of active TII with glomerular involvement, a multivariate analysis, performed in a large retrospective cohort of 59 LuN patients, noted that TII was an independent predictor of ESRD progression and this correlated with serum creatinine level at time of biopsy (OR 2.4, 95% C.I. 1.4, 4.3, p=0.002 for GFR<60 mL/min)(60). Interestingly, in contrast to most current mouse models of lupus and GN, TII has not been shown to be related to levels of complement, double-stranded DNA (dsDNA) or other serum autoantibodies(60, 63). Further studies of TII in animal models have been complicated by an absence of an adequate experimental model, that fully recapitulates the spectrum of tubulointerstitial involvement seen in human patients, such as germinal center formation and other features of tertiary lymphoid neogenesis (TLN)(66–68). To date, characterization of TII in humans has revealed the following common features: the presence of widely interspersed B cells and plasma cells, notably absent from glomeruli(60, 66); co-localization of B and T cells in well-formed aggregates in a majority of biopsy specimens(66); occurrence of discrete immune complex deposits within the tubular basement membrane in 50% of patients(66, 69); and the presence of T follicular helper (TFH) cells in close conjugates with B cells in a significant fraction of patients, which correlated with worse renal function at time of biopsy in a retrospective analysis (mean serum creatinine/GFR by MDRD: 2.3/44.8 mL/min for TFH+ biopsy (n=19) and 1.1/74.8 mL/min, for TFH− biopsy (n=23) patients, respectively; p=0.03)(70). The adaptive nature of the inflammatory response in TII has been further evidenced by clonal relatedness of B cell antibodies, expressed from patient biopsies(66). Analysis of the most common antibody specificities has failed to reveal in situ responses to common nuclear antigens such as dsDNA but, instead, had predominantly cytoplasmic reactivity (11 of 25 expressed antibodies)(71). The majority of these (10 of 11) were recently shown to bind to vimentin – an intermediate filament that is a common structural component of cells(4). In addition, many of the expressed antibodies displayed a degree of poly-reactivity and rheumatoid factor activity – a measure that may reflect the in situ selection environment. Of interest, high titers of these antibodies were almost exclusively seen in biopsies with the highest degree of TII. Anti-vimentin antibodies have been previously described in instances of allograft rejection with high titers predictive of graft failure, and vimentin immunization of a mouse model led to accelerated cardiac rejection(72–75). As such, an anti-vimentin response is unlikely to be specific for lupus but may reflect a general state of deregulated self-tolerance mechanisms in the context of inflammation. An intriguing possibility is whether measurement of anti-vimentin antibodies would be useful as a clinical biomarker of assessing the degree of TII involvement. To date, only anti-C1q antibodies have been prospectively validated to distinguish proliferative from non-proliferating nephritis in human patients but have not been shown to be predictive of clinical course(76).
Table 2.
NIH activity and chronicity index scoring, adapted from (59)
| Index | Glomerular | Tubulointerstitial | Total score |
|---|---|---|---|
| Activity Index | |||
| Maximum Score | 21 | 3 | 24 |
| Individual Components | Cellular Proliferation | Mononuclear Cell Infiltrate | |
| Leukocyte infiltration | |||
| Fibrinoid necrosis/karyorrhexis* | |||
| Cellular crescents* | |||
| Hyaline thrombi/wire loops | |||
| Chronicity Index | |||
| Maximum Score | 6 | 6 | 12 |
| Individual Components | Glomerular sclerosis | Interstitial fibrosis | |
| Fibrous crescents | Tubular atrophy |
Each component in the table above is scored between 0 to 3. Items marked with * are weighted by a factor of 2 (maximum of 6 points each).
Besides the attention paid to features of adaptive immunity in the context of TII, the study of mediators of innate inflammatory responses are also likely to be of great importance. The vast majority of antigen-presenting cells (APCs) in the kidney are not limited to B cells(77). The cortex of normal human kidneys contains a network of myeloid dendritic cells (mDC) as well as fewer numbers of plasmacytoid dendritic cells (pDCs)(78). It also contains macrophages that have similar surface phenotypes, and possibly functions, as dendritic cells (DCs)(79). In murine models, these populations appear to actively maintain tolerance in normal tissue, limit inflammation in response to tissue damage(80–83), drive inflammation in response to ischemia, infection and ureteral obstruction(80, 84–86), and resolve inflammation to allow repair(87, 88). In NZB/NZW mice, kidneys are infiltrated with pro-inflammatory macrophages and DCs(89), and both human and murine lupus is associated with a pattern of in situ mRNA expression, indicative of activated APCs(90). Histologically, human LuN is associated with increased DC infiltration and chemokine expression. An increase in the pDC:mDC ratio has been associated with more severe GN(91, 92). As innate immune responses have previously been more difficult to target therapeutically, this will be an important area of future study. The recent arrival of targeted immunotherapies such as programmed death ligand-1 (PDL-1) blockade(93) may bring about an important revolution in this previously neglected area of treatment of innate-mediated autoimmunity.
A final example of the potential for cooperation between aberrant innate and adaptive responses can be seen in a recent publication, implicating the surprising role of IgE anti-dsDNA antibodies in LuN(94). In this study, the addition of minute quantities of IgE anti-dsDNA antibodies to IgG antibodies with the same specificity was shown to have a potent synergistic effect on pDC production of interferon α. The prevalence of anti-dsDNA IgE antibodies in SLE patients was noted to be markedly elevated as compared to healthy controls and patients with atopic dermatitis (95–97). The authors propose that IgE serves to synergize with and amplify the response to IgG auto-antibodies in an interferon-dependent manner. Indeed, the presence of significant IgE antibody deposits in glomeruli and tubulointerstitium was noted in a majority of biopsies studied, where they co-localized with pDCs, displaying an activated type I inferferon signature. In support of the hypothesis of a potential amplification loop of aberrant in situ responses, the addition of IgE anti-dsDNA antibodies to in vitro pDC:B cell cultures lead to the marked proliferation of B cells, plasma cells, and production of IgM. This data reveals a potential novel mechanism, whereby coordination between the adaptive and innate arms of the immune response creates self-reinforcing pathogenic circuits. They also argue that elucidating precise pathogenic mechanisms is a critical first step in developing appropriate targeted therapies.
Conclusion
While great strides have been made in treating human LuN, a significant percentage of patients still progress to chronic kidney disease and ESRD with conventional treatments. While ACR and EULAR recommendations(14, 15) provide some guidance on traditional induction and maintenance regimens, given the high numbers of LuN patients who do not achieve complete remission with these therapies, current approaches may not be optimal for all patients. Although clinical trials attempt to recruit ethnically diverse patient cohorts, no large RCT to date have been powered to definitively show the superiority of one regimen over another in a particular patient subgroup. Additionally, there is little guidance on how to proceed in patients who are refractory to conventional therapies. Insights into the pathogenesis of LuN and, particularly, the contributions of innate and adaptive arms of the immune system in the context of TII, may become increasingly important in approaching patients with a poor prognosis. The development of an animal model, able to reliably recapitulate the full breadth of human LuN, would represent an important interim step in the development and preclinical validation of appropriate therapeutic targets. In addition to developing new therapeutic agents, future prospective clinical trials need to take TII into consideration as both an important prognostic marker and a central pathogenic mechanism.
Supplementary Material
Supplemental Digital Content 1. Alternate video abstract introduction by two of the authors – VL and KT. mp4
Key points.
The clinical approach to lupus nephritis patient management centers predominantly on glomerular pathology;
The optimal duration of maintenance therapy for lupus nephritis is currently unknown;
Tubulointerstitial inflammation is an important determinant in at least a subset of patients and is tied to faster disease progression to end stage renal disease;
The inflammatory infiltrate possess features of deregulated adaptive and innate immune responses, the future study of which may reveal important clues to prognosis and future therapy development.
Acknowledgments
We wish to thank Andrew Kinloch, Kichul Ko, and Anthony Chang for reviewing and proofreading this manuscript prior to submission.
Financial support:
Vladimir Liarski is supported by NIH NIAMS K08 AR068421.
Marcus Clark is supported by NIH grants U19 AI082724 and AR55646.
Abbreviations
- LuN
lupus nephritis
- SLE
systemic lupus erythematosus
- ESRD
end stage renal disease
- ACR
American College of Rheumatology
- EULAR
European Union League Against Rheumatism
- ISN/RPS
International Society of Nephrology/Renal Pathology Society
- GN
glomerulonephritis
- NIH
National Institute of Health
- GC
glucocorticoid
- IV
intravenous
- CYC
cyclophosphamide
- EL CYC
Euro-Lupus protocol cyclophosphamide
- MMF
mycophenolate mofetil
- AZA
azathioprine
- RCT
randomized controlled trial
- RTX
rituximab
- CR
complete response
- PR
partial response
- OR
overall response
- BAFF
B-cell activating factor
- CTLA4
cytotoxic T-lymphocyte-associated antigen 4
- APRIL
a proliferation-inducing ligand
- TWEAK
TNF-related weak induced of apoptosis
- TACI
transmembrane activator and CAML interactor
- BCMA
B-cell maturation antigen
- MIZ
mizoribine
- LAQ
laquinimod
- TII
tubulointerstitial inflammation
- TLN
tertiary lymphoid neogenesis
- TFH
T follicular helper cells
- dsDNA
double stranded DNA
- APC
antigen presenting cell
- mDC
monocytic dendritic cell
- pDC
plasmacytoid dendritic cell
- DC
dendritic cell
- PDL-1
programmed death ligand 1
Footnotes
Alternate video abstract introduction (see Video, Supplemental Digital Content 1, with introduction by two of the authors – VL and KT).
Conflicts of Interest and Source of Funding:
The authors have no conflicts of interest or sources of funding to declare
Annotated References
- 1.Mok C, Tang SS. Incidence and predictors of renal disease in Chinese patients with systemic lupus erythematosus. Am J Med. 2004;117(10):791–5. doi: 10.1016/j.amjmed.2004.04.029. [DOI] [PubMed] [Google Scholar]
- 2.Cervera R, Khamashta MA, Font J, Sebastiani GD, Gil A, Lavilla P, Mejia JC, Aydingtug AO, Chwalinska-Sadowska H, de Ramon E, Fernandez-Nebro A, Galeazzi M, Valen M, Mathieu A, Houssiau F, Caro N, Alba P, Ramos-Casals M, Ingelmo M, Hughes GR European Working Party on Systemic Lupus Erythematosus. Morbidity and mortality in systemic lupus erythematosus during a 10-year period: a comparison of early and late manifestations in a cohort of 1,000 patients. Medicine (Baltimore) 2003;82(5):299–308. doi: 10.1097/01.md.0000091181.93122.55. [DOI] [PubMed] [Google Scholar]
- 3.Bernatsky S, Boivin JF, Joseph L, Manzi S, Ginzler E, Gladman DD, Urowitz M, Fortin PR, Petri M, Barr S, Gordon C, Bae SC, Isenberg D, Zoma A, Aranow C, Dooley MA, Nived O, Sturfelt G, Steinsson K, Alarcon G, Senecal JL, Zummer M, Hanly J, Ensworth S, Pope J, Edworthy S, Rahman A, Sibley J, El-Gabalawy H, McCarthy T, St Pierre Y, Clarke A, Ramsey-Goldman R. Mortality in systemic lupus erythematosus. Arthritis and rheumatism. 2006;54(8):2550–7. doi: 10.1002/art.21955. [DOI] [PubMed] [Google Scholar]
- 4.McLaughlin J, Bombardier C, Farewell VT, Gladman DD, Urowitz MB. Kidney biopsy in systemic lupus erythematosus. III. Survival analysis controlling for clinical and laboratory variables. Arthritis and rheumatism. 1994;37(4):559–67. doi: 10.1002/art.1780370417. [DOI] [PubMed] [Google Scholar]
- 5.Cook R, Gladman DD, Pericak D, Urositz MB. Prediction of short term mortality in systemic lupus erythematosus with time dependent measures of disease activity. J Rheumatol. 2000;27(8):1892–5. [PubMed] [Google Scholar]
- 6.Flanc R, Roberts MA, Strippoli GF, Chadban SJ, Kerr PG, Atkins RC. Treatment for lupus nephritis. Cochrane Database Syst Rev. 2004;2004(1):CD0002922. doi: 10.1002/14651858.CD002922.pub2. [DOI] [PubMed] [Google Scholar]
- 7.Ginzler EM, Dooley MA, Aranow C, Kim MY, Buyon J, Merrill JT, et al. Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis. N Engl J Med. 2005;353(21):2219–28. doi: 10.1056/NEJMoa043731. [DOI] [PubMed] [Google Scholar]
- 8.Appel GB, Contreras G, Dooley MA, Ginzler EM, Isenberg D, Jayne D, et al. Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J Am Soc Nephrol. 2009;20(5):1103–12. doi: 10.1681/ASN.2008101028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Korbet S, Schwartz MM, Evans J, Lewis EJ. Severe lupus nephritis:racial differences in presentation and outcome. J Am Soc Nephrol. 2007;18:244–54. doi: 10.1681/ASN.2006090992. [DOI] [PubMed] [Google Scholar]
- 10.Donadio JJ, Hart GM, Bergstralh EJ, Holley KE. Prognostic determinants in lupus nephritis: a long-term clinicopathologic study. Lupus. 1995;4(2):109–15. doi: 10.1177/096120339500400206. [DOI] [PubMed] [Google Scholar]
- 11.Neumann K, Wallace DJ, Azen C, Nessim S, Fichman M, Metzger AL, Klinenberg JR. Lupus in the 1980s: III. Influence of clinical variables, biopsy, and treatment on the outcome in 150 patients with lupus nephritis seen at a single center. Semin Arthritis Rheum. 1995;25(1):47–55. doi: 10.1016/s0049-0172(95)80017-4. [DOI] [PubMed] [Google Scholar]
- 12.Austin Hr, Klippel JH, Balow JE, le Riche NG, Steinberg AD, Plotz PH, Decker JL. Therapy of lupus nephritis. Controlled trial of prednisone and cytotoxic drugs. N Engl J Med. 1986;314(10):614–9. doi: 10.1056/NEJM198603063141004. [DOI] [PubMed] [Google Scholar]
- 13.Ong L, Hooi LS, Lim TO, Goh BL, Ahmad G, Ghazalli R, Teo SM, Wong HS, Tan SY, Shaariah W, Tan CC, Morad Z. Randomized controlled trial of pulse intravenous cyclophosphamide versus mycophenolate mofetil in the induction therapy of proliferative lupus nephritis. Nephrology (Carlton) 2005;10(5):504–10. doi: 10.1111/j.1440-1797.2005.00444.x. [DOI] [PubMed] [Google Scholar]
- 14.Hahn BH, McMahon MA, Wilkinson A, Wallace WD, Daikh DI, Fitzgerald JD, et al. American College of Rheumatology guidelines for screening, treatment, and management of lupus nephritis. Arthritis Care Res (Hoboken) 2012;64(6):797–808. doi: 10.1002/acr.21664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bertsias GK, Tektonidou M, Amoura Z, Aringer M, Bajema I, Berden JH, 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(11):1771–82. doi: 10.1136/annrheumdis-2012-201940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Weening JJ, D’Agati VD, Schwartz MM, Seshan SV, Alpers CE, Appel GB, et al. The classification of glomerulonephritis in systemic lupus erythematosus revisited. J Am Soc Nephrol. 2004;15(2):241–50. doi: 10.1097/01.asn.0000108969.21691.5d. [DOI] [PubMed] [Google Scholar]
- 17.Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, de Garrido Ed ER, Danieli MG, et al. Immunosuppressive therapy in lupus nephritis: the Euro-Lupus Nephritis Trial, a randomized trial of low-dose versus high-dose intravenous cyclophosphamide. Arthritis and rheumatism. 2002;46(8):2121–31. doi: 10.1002/art.10461. [DOI] [PubMed] [Google Scholar]
- 18.Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, de Ramon Garrido E, Danieli MG, et al. The 10-year follow-up data of the Euro-Lupus Nephritis Trial comparing low-dose and high-dose intravenous cyclophosphamide. Ann Rheum Dis. 2010;69(1):61–4. doi: 10.1136/ard.2008.102533. [DOI] [PubMed] [Google Scholar]
- 19.Chan TM, Li FK, Tang CS, Wong RW, Fang GX, Ji YL, et al. Efficacy of mycophenolate mofetil in patients with diffuse proliferative lupus nephritis. Hong Kong-Guangzhou Nephrology Study Group. N Engl J Med. 2000;343(16):1156–62. doi: 10.1056/NEJM200010193431604. [DOI] [PubMed] [Google Scholar]
- 20.Mok CC, Ying KY, Ng WL, Lee KW, To CH, Lau CS, et al. Long-term outcome of diffuse proliferative lupus glomerulonephritis treated with cyclophosphamide. Am J Med. 2006;119(4):355, e25–33. doi: 10.1016/j.amjmed.2005.08.045. [DOI] [PubMed] [Google Scholar]
- 21.Mok CC. Towards new avenues in the management of lupus glomerulonephritis. Nat Rev Rheumatol. 2016 doi: 10.1038/nrrheum.2015.174. [DOI] [PubMed] [Google Scholar]
- 22.Dooley MA, Jayne D, Ginzler EM, Isenberg D, Olsen NJ, Wofsy D, et al. Mycophenolate versus azathioprine as maintenance therapy for lupus nephritis. N Engl J Med. 2011;365(20):1886–95. doi: 10.1056/NEJMoa1014460. [DOI] [PubMed] [Google Scholar]
- 23.Houssiau FA, D’Cruz D, Sangle S, Remy P, Vasconcelos C, Petrovic R, et al. Azathioprine versus mycophenolate mofetil for long-term immunosuppression in lupus nephritis: results from the MAINTAIN Nephritis Trial. Ann Rheum Dis. 2010;69(12):2083–9. doi: 10.1136/ard.2010.131995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24**.Tamirou F, D’Cruz D, Sangle S, Remy P, Vasconcelos C, Fiehn C, 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(3):526–31. doi: 10.1136/annrheumdis-2014-206897. This is a 10 year follow-up trial of maintenance therapy for lupus nephritis that showed equal efficacy of azathioprine and mycophenolate. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Austin HA, 3rd, Illei GG, Braun MJ, Balow JE. Randomized, controlled trial of prednisone, cyclophosphamide, and cyclosporine in lupus membranous nephropathy. J Am Soc Nephrol. 2009;20(4):901–11. doi: 10.1681/ASN.2008060665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Radhakrishnan J, Moutzouris DA, Ginzler EM, Solomons N, Siempos II, Appel GB. Mycophenolate mofetil and intravenous cyclophosphamide are similar as induction therapy for class V lupus nephritis. Kidney Int. 2010;77(2):152–60. doi: 10.1038/ki.2009.412. [DOI] [PubMed] [Google Scholar]
- 27.Pasquali S, Banfi G, Zucchelli A, Moroni G, Ponticelli C, Zucchelli P. Lupus membranous nephropathy: long-term outcome. Clin Nephrol. 1993;39(4):175–82. [PubMed] [Google Scholar]
- 28.Mok CC, Lau CS, Chan TM, Wong RW. Clinical characteristics and outcome of southern Chinese males with systemic lupus erythematosus. Lupus. 1999;8(3):188–96. doi: 10.1191/096120399678847605. [DOI] [PubMed] [Google Scholar]
- 29.Ward MM. Recent clinical trials in lupus nephritis. Rheum Dis Clin North Am. 2014;40(3):519–35. ix. doi: 10.1016/j.rdc.2014.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chen W, Tang X, Liu Q, Chen W, Fu P, Liu F, et al. Short-term outcomes of induction therapy with tacrolimus versus cyclophosphamide for active lupus nephritis: A multicenter randomized clinical trial. Am J Kidney Dis. 2011;57(2):235–44. doi: 10.1053/j.ajkd.2010.08.036. [DOI] [PubMed] [Google Scholar]
- 31*.Mok CC, Ying KY, Yim CW, Siu YP, Tong KH, To CH, et al. Tacrolimus versus mycophenolate mofetil for induction therapy of lupus nephritis: a randomised controlled trial and long-term follow-up. Ann Rheum Dis. 2016;75(1):30–6. doi: 10.1136/annrheumdis-2014-206456. This is the largest trial to date comparing tacrolimus with mycophenolate for treatment of lupus nephritis that showed non-inferiority at 6 months. [DOI] [PubMed] [Google Scholar]
- 32**.Liu Z, Zhang H, Liu Z, Xing C, Fu P, Ni Z, et al. Multitarget therapy for induction treatment of lupus nephritis: a randomized trial. Annals of internal medicine. 2015;162(1):18–26. doi: 10.7326/M14-1030. This is the first trial to show that combination therapy with tacrolimus and mycophenolate was superior to IV cyclophosphamide alone after 6 months of treatment. [DOI] [PubMed] [Google Scholar]
- 33.Zhang X, Ji L, Yang L, Tang X, Qin W. The effect of calcineurin inhibitors in the induction and maintenance treatment of lupus nephritis: a systematic review and meta-analysis. Int Urol Nephrol. 2016 doi: 10.1007/s11255-015-1201-z. [DOI] [PubMed] [Google Scholar]
- 34.Pepper R, Griffith M, Kirwan C, Levy J, Taube D, Pusey C, et al. Rituximab is an effective treatment for lupus nephritis and allows a reduction in maintenance steroids. Nephrol Dial Transplant. 2009;24(12):3717–23. doi: 10.1093/ndt/gfp336. [DOI] [PubMed] [Google Scholar]
- 35.Gunnarsson I, Sundelin B, Jonsdottir T, Jacobson SH, Henriksson EW, van Vollenhoven RF. Histopathologic and clinical outcome of rituximab treatment in patients with cyclophosphamide-resistant proliferative lupus nephritis. Arthritis and rheumatism. 2007;56(4):1263–72. doi: 10.1002/art.22505. [DOI] [PubMed] [Google Scholar]
- 36.Terrier B, Amoura Z, Ravaud P, Hachulla E, Jouenne R, Combe B, et al. Safety and efficacy of rituximab in systemic lupus erythematosus: results from 136 patients from the French AutoImmunity and Rituximab registry. Arthritis and rheumatism. 2010;62(8):2458–66. doi: 10.1002/art.27541. [DOI] [PubMed] [Google Scholar]
- 37.Marenco JL, Fernandez-Nebro A, en nombre de L. Use of rituximab in the treatment of lupus patients. The LESIMAB database. Reumatol Clin. 2010;6(Suppl 2):28–33. doi: 10.1016/j.reuma.2010.06.002. [DOI] [PubMed] [Google Scholar]
- 38.Melander C, Sallee M, Trolliet P, Candon S, Belenfant X, Daugas E, et al. Rituximab in severe lupus nephritis: early B-cell depletion affects long-term renal outcome. Clin J Am Soc Nephrol. 2009;4(3):579–87. doi: 10.2215/CJN.04030808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Vigna-Perez M, Hernandez-Castro B, Paredes-Saharopulos O, Portales-Perez D, Baranda L, Abud-Mendoza C, et al. Clinical and immunological effects of Rituximab in patients with lupus nephritis refractory to conventional therapy: a pilot study. Arthritis Res Ther. 2006;8(3):R83. doi: 10.1186/ar1954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. Arthritis and rheumatism. 2012;64(4):1215–26. doi: 10.1002/art.34359. [DOI] [PubMed] [Google Scholar]
- 41.Moroni G, Raffiotta F, Trezzi B, Giglio E, Mezzina N, Del Papa N, et al. Rituximab vs mycophenolate and vs cyclophosphamide pulses for induction therapy of active lupus nephritis: a clinical observational study. Rheumatology (Oxford) 2014;53(9):1570–7. doi: 10.1093/rheumatology/ket462. [DOI] [PubMed] [Google Scholar]
- 42.Navarra SV, Guzman RM, Gallacher AE, Hall S, Levy RA, Jimenez RE, et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377(9767):721–31. doi: 10.1016/S0140-6736(10)61354-2. [DOI] [PubMed] [Google Scholar]
- 43.Furie R, Petri M, Zamani O, Cervera R, Wallace DJ, Tegzova D, et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis and rheumatism. 2011;63(12):3918–30. doi: 10.1002/art.30613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dooley MA, Houssiau F, Aranow C, D’Cruz DP, Askanase A, Roth DA, et al. Effect of belimumab treatment on renal outcomes: results from the phase 3 belimumab clinical trials in patients with SLE. Lupus. 2013;22(1):63–72. doi: 10.1177/0961203312465781. [DOI] [PubMed] [Google Scholar]
- 45.Petri M, Kim MY, Kalunian KC, Grossman J, Hahn BH, Sammaritano LR, et al. Combined oral contraceptives in women with systemic lupus erythematosus. N Engl J Med. 2005;353(24):2550–8. doi: 10.1056/NEJMoa051135. [DOI] [PubMed] [Google Scholar]
- 46.Group AT. Treatment of lupus nephritis with abatacept: the Abatacept and Cyclophosphamide Combination Efficacy and Safety Study. Arthritis Rheumatol. 2014;66(11):3096–104. doi: 10.1002/art.38790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Furie R, Nicholls K, Cheng TT, Houssiau F, Burgos-Vargas R, Chen SL, et al. Efficacy and safety of abatacept in lupus nephritis: a twelve-month, randomized, double-blind study. Arthritis Rheumatol. 2014;66(2):379–89. doi: 10.1002/art.38260. [DOI] [PubMed] [Google Scholar]
- 48.Gross JA, Johnston J, Mudri S, Enselman R, Dillon SR, Madden K, et al. TACI and BCMA are receptors for a TNF homologue implicated in B-cell autoimmune disease. Nature. 2000;404(6781):995–9. doi: 10.1038/35010115. [DOI] [PubMed] [Google Scholar]
- 49.Ginzler EM, Wax S, Rajeswaran A, Copt S, Hillson J, Ramos E, et al. Atacicept in combination with MMF and corticosteroids in lupus nephritis: results of a prematurely terminated trial. Arthritis Res Ther. 2012;14(1):R33. doi: 10.1186/ar3738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Renaudineau Y, Devauchelle-Pensec V, Hanrotel C, Pers JO, Saraux A, Youinou P. Monoclonal anti-CD20 antibodies: mechanisms of action and monitoring of biological effects. Joint Bone Spine. 2009;76(5):458–63. doi: 10.1016/j.jbspin.2009.03.010. [DOI] [PubMed] [Google Scholar]
- 51.Mysler EF, Spindler AJ, Guzman R, Bijl M, Jayne D, Furie RA, et al. Efficacy and safety of ocrelizumab in active proliferative lupus nephritis: results from a randomized, double-blind, phase III study. Arthritis and rheumatism. 2013;65(9):2368–79. doi: 10.1002/art.38037. [DOI] [PubMed] [Google Scholar]
- 52.Stohl W. Future prospects in biologic therapy for systemic lupus erythematosus. Nat Rev Rheumatol. 2013;9(12):705–20. doi: 10.1038/nrrheum.2013.136. [DOI] [PubMed] [Google Scholar]
- 53.Stohl W, Merrill JT, Looney RJ, Buyon J, Wallace DJ, Weisman MH, et al. Treatment of systemic lupus erythematosus patients with the BAFF antagonist “peptibody” blisibimod (AMG 623/A-623): results from randomized, double-blind phase 1a and phase 1b trials. Arthritis Res Ther. 2015;17:215. doi: 10.1186/s13075-015-0741-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ishikawa H. Mizoribine and mycophenolate mofetil. Curr Med Chem. 1999;6(7):575–97. [PubMed] [Google Scholar]
- 55.Feng X, Gu F, Chen W, Liu Y, Wei H, Liu L, et al. Mizoribine versus mycophenolate mofetil or intravenous cyclophosphamide for induction treatment of active lupus nephritis. Chin Med J (Engl) 2014;127(21):3718–23. [PubMed] [Google Scholar]
- 56.Peng L, Oganesyan V, Wu H, Dall’Acqua WF, Damschroder MM. Molecular basis for antagonistic activity of anifrolumab, an anti-interferon-alpha receptor 1 antibody. MAbs. 2015;7(2):428–39. doi: 10.1080/19420862.2015.1007810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Morehouse C, Chang L, Wang L, Brohawn P, Uesa S, Illei G. Target modulation of a type i interferon (IFN) gene signature with sifalimumab or anifrolumab in systemic lupus erythematosus (SLE) patients in two open label phase 2 Japanese trials. Arthritis and rheumatism. 2014;66:719. [Google Scholar]
- 58.Jayne D, Appel G, Chan TM, Barkay H, Weiss R, Wofsy D. LB0003 A Randomized Controlled Study of Laquinimod in Active Lupus Nephritis Patients in Combination with Standard of Care. Ann Rheum Dis. 2013;72(Suppl 3):A164. [Google Scholar]
- 59.Austin HA, 3rd, Muenz LR, Joyce KM, Antonovych TA, Kullick ME, Klippel JH, et al. Prognostic factors in lupus nephritis. Contribution of renal histologic data. Am J Med. 1983;75(3):382–91. doi: 10.1016/0002-9343(83)90338-8. [DOI] [PubMed] [Google Scholar]
- 60.Hsieh C, Chang A, Brandt D, Guttikonda R, Utset TO, Clark MR. Predicting outcomes of lupus nephritis with tubulointerstitial inflammation and scarring. Arthritis Care Res (Hoboken) 2011;63(6):865–74. doi: 10.1002/acr.20441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Esdaile JM, Levinton C, Federgreen W, Hayslett JP, Kashgarian M. The clinical and renal biopsy predictors of long-term outcome in lupus nephritis: a study of 87 patients and review of the literature. The Quarterly journal of medicine. 1989;72(269):779–833. [PubMed] [Google Scholar]
- 62.Park MH, D’Agati V, Appel GB, Pirani CL. Tubulointerstitial disease in lupus nephritis: relationship to immune deposits, interstitial inflammation, glomerular changes, renal function, and prognosis. Nephron. 1986;44:309–19. doi: 10.1159/000184012. [DOI] [PubMed] [Google Scholar]
- 63.Hill GS, Delahousse M, Nochy D, Tomkiewicz E, Remy P, Mignon F, et al. A new morphologic index for the evaluation of renal biopsies in lupus nephritis. Kid Int. 2000;58:1160–73. doi: 10.1046/j.1523-1755.2000.00272.x. [DOI] [PubMed] [Google Scholar]
- 64.Schwartz M, Fennell JS, Lewis EJ. Pathologic changes in the renal tubule in systemic lupus erythematosus. Hum Pathol. 1982;13:534–47. doi: 10.1016/s0046-8177(82)80268-2. [DOI] [PubMed] [Google Scholar]
- 65.Bohle A, Wehrmann M, Bogenschutz O, Batz C, Vogl W, Schmitt H, et al. The long-term prognosis of the primary glomerulonephritides. A morphological and clinical analysis of 1747 cases. Pathol Res Pract. 1992;188:908–24. doi: 10.1016/s0344-0338(11)80252-9. [DOI] [PubMed] [Google Scholar]
- 66.Chang A, Henderson SG, Liu N, Guttikonda R, Hsieh C, Utset TO, et al. In situ B cell-mediated immune responses and tubulointerstitial inflammation in human lupus nephritis. Journal of immunology. 2011;186(3):1849–60. doi: 10.4049/jimmunol.1001983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Perry D, Sang A, Yin Y, Zheng YY, Morel L. Murine models of systemic lupus erythematosus. J Biomed Biotechnol. 2011;2011:271694. doi: 10.1155/2011/271694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Steinmetz OM, Velden J, Kneissler U, Marx M, Klein A, Helmchen U, et al. Analysis and classification of B-cell infiltrates in lupus and ANCA-associated vasculitis. Kid Int. 2008 doi: 10.1038/ki.2008.191. epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 69.Satoskar AA, Brodsky SV, Nadasdy G, Bott C, Rovin B, Hebert L, et al. Discrepancies in glomerular and tubulointerstitial/vascular immune complex IgG subclasses in lupus nephritis. Lupus. 2011;20(13):1396–403. doi: 10.1177/0961203311416533. [DOI] [PubMed] [Google Scholar]
- 70.Liarski VM, Kaverina N, Chang A, Brandt D, Yanez D, Talasnik L, et al. Cell distance mapping identifies functional T follicular helper cells in inflamed human renal tissue. Science translational medicine. 2014;6(230):230ra46. doi: 10.1126/scitranslmed.3008146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kinloch AJ, Chang A, Ko K, Henry Dunand CJ, Henderson S, Maienschein-Cline M, et al. Vimentin is a dominant target of in situ humoral immunity in human lupus tubulointerstitial nephritis. Arthritis Rheumatol. 2014;66(12):3359–70. doi: 10.1002/art.38888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Jurcevic S, Ainsworth ME, Pomerance A, Smith JD, Robinson DR, Dunn MJ, et al. Antivimentin antibodies are an independent predictor of transplant-associated coronary artery disease after cardiac transplantation. Transplantation. 2001;71:886–92. doi: 10.1097/00007890-200104150-00011. [DOI] [PubMed] [Google Scholar]
- 73.Nath DS, Ilias BG, Tiriveedhi V, Alur C, Phelan D, Eward GA, et al. Characterization of immune responses to cardiac self-antigens myosin and vimentin in human cardiac allograft recipients with antibody-mediated rejection and cardiac allograft vasculopathy. J Heart Lung Transplant. 2010;29:1277–85. doi: 10.1016/j.healun.2010.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Carter V, Shenton BK, Jacques B, Turner D, Talbot D, Gupta A. Anti-vimentin antibody detection in recipients of heart-beating and non-heart beating donor kidneys. Transplant Proc. 2005;8:3269–71. doi: 10.1016/j.transproceed.2005.09.062. [DOI] [PubMed] [Google Scholar]
- 75.Mahesh B, Leong JS, McCormak A, Sarathchandra P, Holder A, Rose ML. Autoantibodies to vimentin cause accelerated rejection of cardiac allografts. The American journal of pathology. 2007;170:1415–27. doi: 10.2353/ajpath.2007.060728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76**.Moroni G, Quaglini S, Radice A, Trezzi B, Raffiotta F, Messa P, et al. 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. doi: 10.1155/2015/106904. This is one of the few large prospective trials, confirming the association of anti-C1q antibodies with proliferative as compared to non-proliferative lupus nephritis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Johnson RJ, Feehally J, Floege J. Comprehensive clinical nephrology. 5. Philadelphia, PA: Elsevier/Saunders; 2015. p. xx.p. 1299. [Google Scholar]
- 78.Woltman AM, de Fijter JW, Zuidwijk K, Vlug AG, Bajema IM, van der Kooij SW, et al. Quantification of dendritic cell subsets in human renal tissue under normal and pathological conditions. Kid Int. 2007;71:1001–8. doi: 10.1038/sj.ki.5002187. [DOI] [PubMed] [Google Scholar]
- 79.Nelson PJ, Rees AJ, Griffin MD, Hughes J, Kurts C, Duffield J. The renal mononuclear phagocytic system. J Am Soc Nephrol. 2012;23:194–203. doi: 10.1681/ASN.2011070680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Dong X, Swaminathan S, Bachman LA, Croatt AJ, Nath KA, Griffin MD. Antigen presentation by dendritic cells in renal lymph nodes is linked to systemic and local injury to the kidney. Kidney Int. 2005;68:1096–108. doi: 10.1111/j.1523-1755.2005.00502.x. [DOI] [PubMed] [Google Scholar]
- 81.Lukacs-Kornek V, Burgdorf S, Diehl L, Specht S, Kornek M, Kurts C. The kidney-renal lymph node-system contributes to cross-tolerance against innocuous circulating antigen. Journal of immunology. 2008;180:706–15. doi: 10.4049/jimmunol.180.2.706. [DOI] [PubMed] [Google Scholar]
- 82.Coates PT, Duncan FJ, Colvin GL, Wang Z, Zhahorchak AF, Shufesky WJ, et al. In vivo-mobilized kidney dendritic cells are functionally immature, subvert alloreactive T-cell responses, and prolong organ allograft survival. Transplantation. 2004;77:1080–9. doi: 10.1097/01.tp.0000122183.60680.c9. [DOI] [PubMed] [Google Scholar]
- 83.Scholz JL, Lukas-Kornek V, Engel DR, Specht S, Kiss E, Eitner F, et al. Renal dendritic cells stimulate IL-10 production and attenuate nephrotoxic nephritis. J Am Soc Nephrol. 2008;19:527–37. doi: 10.1681/ASN.2007060684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Dong X, Bachman LA, Miller MN, Nath KA, Griffin MD. Dendritic cells facilitate accumulation of IL-17 T cells in the kidney following acute renal obstruction. Kidney Int. 2008;74:1294–1309. doi: 10.1038/ki.2008.394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Dong X, Swarminathan S, Bachman LA, Croatt AJ, Nath KA, Griffin MD. Resident dendritic cells are the predominant TNF-secreting cell in early renal ischemia-reperfusion injury. Kid Int. 2007;71:619–28. doi: 10.1038/sj.ki.5002132. [DOI] [PubMed] [Google Scholar]
- 86.Tittel AP, Heuser C, Ohliger C, Knoller PA, Engel DR, Kurts C. Kideny dendritic cells induce innate immunity against bacterial pyelonephritis. J Am Soc Nephrol. 2011;22:1435–41. doi: 10.1681/ASN.2010101072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Cao Q, Wang Y, Zheng D, Sun Y, Wang Y, Lee VWS, et al. IL-10/TGF-beta-modified macrophages induce regulatory T cells and protect against adriamycin nephrosis. J Am Soc Nephrol. 2010;21:933–942. doi: 10.1681/ASN.2009060592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Lee S, Huen S, Nishio H, Nishio S, Lee HK, Choi BS, et al. Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol. 2011;22:317–26. doi: 10.1681/ASN.2009060615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bethunaickan R, Berthier CC, Ramanujam M, Sahu R, Zhang W, Sun Y, et al. A unique hybrid renal mononuclear phagocyte activation phenotype in murine systemic lupus erythematosus nephritis. Journal of immunology. 2011;186:4994–5003. doi: 10.4049/jimmunol.1003010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Berthier CC, Bethunaickan R, Gonzalez-Riveria T, Nair V, Ramanujam M, Zhang W, et al. Cross-species transcriptional network analysis defines shared inflammatory responses in murine and human lupus nephritis. Journal of immunology. 2012;189:988–1001. doi: 10.4049/jimmunol.1103031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Fiore N, Castellano G, Blasi A, Capobianco C, Loverre A, Montinaro V, et al. Immature myeloid and plasmacytoid dendritic cells infiltrate renal tubulointerstitium in patients with lupus nephritis. Mol Immunol. 2008;45:259–65. doi: 10.1016/j.molimm.2007.04.029. [DOI] [PubMed] [Google Scholar]
- 92.De Palma G, Castellano G, Del Prete A, Sozzani S, Fiore N, Loverre A, et al. The possible role of ChemR23/Chemerin axis in the recruitment of dendritic cells in lupus nephritis. Kid Int. 2011;79:1228–35. doi: 10.1038/ki.2011.32. [DOI] [PubMed] [Google Scholar]
- 93.Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest. 2015;125(9):3384–91. doi: 10.1172/JCI80011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94*.Henault J, Riggs JM, Karnell JL, Liarski VM, Li J, Shirinian L, et al. Self-reactive IgE exacerbates interferon responses associated with autoimmunity. Nature immunology. 2016;17(2):196–203. doi: 10.1038/ni.3326. This is the first trial, showing an association of IgE dsDNA antibodies with human lupus and their relation with renal inflammation, as mediated by increased type I inferferon production by resident plasmacytoid dendritic cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Elkayam O, Tamir R, Pick AI, Wysenbeek A. Serum IgE concentrations, disease activity, and atopic disorders in systemic lupus erythematosus. Allergy. 1995;50(1):94–6. [PubMed] [Google Scholar]
- 96.Morton S, Palmer B, Muir K, Powell RJ. IgE and non-IgE mediated allergic disorders in systemic lupus erythematosus. Ann Rheum Dis. 1998;57(11):660–3. doi: 10.1136/ard.57.11.660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Wozniacka A, Sysa-Jedrzejowska A, Robak E, Samochocki Z, Zak-Prelich M. Allergic diseases, drug adverse reactions and total immunoglobulin E levels in lupus erythematosus patients. Mediators Inflamm. 2003;12(2):95–9. doi: 10.1080/0962935031000097709. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental Digital Content 1. Alternate video abstract introduction by two of the authors – VL and KT. mp4
