You are on page 1of 12

26

Lupus Nephritis
Shikha Wadhwani, David Jayne, Brad H. Rovin

DEFINITION high frequency of autoantibodies and other autoimmune disorders in


uninvolved family members of SLE patients.11 A Taiwanese study found
Lupus nephritis (LN), the prototypical immune complex glomerulo- that the relative risk for lupus in families of those with SLE was 316
nephritis (GN), is common and serious in patients with systemic lupus for twins, 24 for siblings, 11 for parents, 14 for children of patients,
erythematosus (SLE). SLE is defined by clinical and laboratory features and 4.4 for nonrelated spouses, leading to estimates that heritability
as outlined in the 1997 American College of Rheumatology (ACR) contributed 44% to the risk for SLE, shared environmental exposures
criteria for the diagnosis of SLE (Box 26.1). The Systemic Lupus Inter- contributed 26%, and nonshared environmental exposures 30%.12
national Collaborating Clinics (SLICC) criteria introduced in 2012 (see Homozygous deficiency of early complement cascade components
Box 26.1) have shown greater diagnostic sensitivity and fewer misclas- (C1q, C2, C4) carries a high risk for development of SLE; this is also
sifications than the ACR criteria.1,2 The SLICC criteria include biopsy- true for certain Fc γ RIII receptor polymorphisms. Genome-wide asso-
proven nephritis compatible with LN in the presence of either antinuclear ciation studies have identified over 100 loci associated with an increased
antibody (ANA) or anti–double-stranded (ds)DNA antibody as sufficient risk for SLE, although the individual contribution of each gene to overall
evidence for the diagnosis of SLE, but eliminated dipstick-positive risk is low. Genetic risk scores are lowest in Europeans, higher in Asians,
albuminuria as sufficient to confirm kidney involvement in LN. The and highest in those of African ancestry.13 Genes involved in lupus risk
SLICC criteria may influence the apparent incidence of LN. include those affecting B cell signaling, neutrophil function, interferon
regulation, immune complex clearance, and Toll-like receptors.11 A
disproportionate number of transcription factors have been identified,
EPIDEMIOLOGY suggesting an important contribution of gene dysregulation to lupus.14
The incidence and prevalence of lupus and LN are influenced by age, HLA genes are strongly associated with lupus risk, at least in European
gender, ethnicity, geographical region, diagnostic criteria used, and populations.15 A meta-analysis of HLA class II alleles associated with
method of ascertainment, but across populations, clinically important SLE identified four DR allele families as conferring increased suscep-
kidney disease (Table 26.1) will occur in about 50% of patients with tibility or resistance to the development of LN when compared with
SLE.3,4 The peak incidence of lupus is between the ages of 15 and 45 healthy controls.16 This study involved mostly White and Asian patients,
years, with women outnumbering men 8 to 15 : 1. Female predominance and the findings may not be applicable to other patient groups. Inde-
is less pronounced in children and older individuals. Among patients pendent of causation, other polymorphisms in genes such as MYH-
with lupus, LN affects both sexes equally, is more severe in children 9, ACE, TNIP-1, and APOL-1 (in Blacks) confer a worse prognosis
and men, and is less so in older adults. The incidence of LN is about for LN.
30% in White, 60% in Black and Hispanic, and 40% to 80% in Asian External environmental factors such as ultraviolet light exposure
patients with SLE.3,5 The higher frequency of LN in Black patients with and smoking play a role in the onset and exacerbation of SLE and LN.17
lupus in the United States persists after adjusting for socioeconomic Viral infections may be triggers for lupus, but conclusive evidence for
status.6 Black and Hispanic patients with SLE develop LN earlier and a viral pathogenesis of SLE or LN has yet to be produced. Exposure to
have worse outcomes, including death and end-stage renal disease certain medications (e.g., procainamide, hydralazine, quinidine, and
(ESRD), than White patients. The prevalence of LN in a U.S. Medicaid anti-TNF biologics) has been linked to SLE or SLE-like syndromes, but
population was 31 in 100,000.6 LN occurs infrequently in such individuals. The internal environment
Approximately 10% of patients with LN will develop ESRD3; however, also appears to be relevant given the strong female predominance of
this depends on histologic class. The risk for ESRD over 15 years was SLE, disease exacerbations during or shortly after pregnancy, and the
found to be as high as 44% in class 4 LN.4 effects of hormone treatment and ablation in animal models of LN.
SLE patients with LN die earlier than those without nephritis and
have a standardized mortality ratio of 6 to 6.8 versus 2.4 in lupus Autoimmunity in Systemic Lupus Erythematosus
without renal involvement.7,8 This increases to 14 for those with chronic Patients with SLE typically develop multiple autoantibodies, many of
kidney disease and 63 for those with ESRD.9 However, if LN remission which are directed against nucleic acids and proteins involved in tran-
is achieved through treatment, 10-year survival doubles to 95%.10 scription and translation, such as nucleosomes (DNA-histone), chromatin
antigens, and small nuclear and cytoplasmic ribonuclear proteins.18 In
ETIOLOGY AND PATHOGENESIS the early stages of disease, clearance of apoptotic cells is impaired and
nuclear autoantigens released from these cells stimulate expression of
Genetics and Environment interferon-α (IFN-α), which facilitates the generation of antigen-
A genetic predisposition to SLE is supported by disease clustering in presenting cells, promotes the differentiation of autoreactive B cells
families, twin concordance, racial differences in susceptibility, and the into plasma cells, and fosters the development of T helper cells. Antigen

306
CHAPTER 26 Lupus Nephritis 307

BOX 26.1 American College of Rheumatology (ACR) and Systemic Lupus International
Collaborating Clinics (SLICC) Criteria for the Diagnosis of Lupus
ACR Criteria SLICC ACR Criteria
Presence (cumulative) of four or more of the following: Presence (cumulative) of four or more of the following (with at least 1 clinical and 1
immunologic criteria) OR biopsy-proven lupus nephritis with positive ANA or anti-dsDNA
Clinical Criteria Immunologic Criteria
1. Malar rash 1. Acute OR subacute cutaneous lupus 1. Positive ANA
2. Discoid rash 2. Chronic cutaneous lupus 2. Positive anti-dsDNA antibody
3. Photosensitivity 3. Nonscarring alopecia 3. Positive anti-Sm antibody
4. Oral or nasopharyngeal ulcers 4. Oral OR nasal ulcers 4. Positive antiphospholipid antibody (includes
presence of a lupus anticoagulant,
false-positive RPR, anticardiolipin antibody
or anti-β2 glycoprotein antibody)
5. Nonerosive arthritis (involving ≥2 joints, 5. Synovitis ≥2 joints (swelling or 5. Low complement (C3, C4, or CH50)
characterized by tenderness, swelling, or effusion) effusion) OR tenderness in 2 or more
joints and ≥30 min of morning stiffness
6. Pleuritis or pericarditis 6. Serositis 6. Direct Coombs test (in the absence of
hemolytic anemia)
7. Renal disease (proteinuria >500 mg/day OR 3+ by 7. Renal disease (red blood cell casts OR
dipstick OR cellular casts) proteinuria ≥500 mg/day on 24-hr urine
collection OR spot ratio of urine
protein to creatinine ratio ≥0.5)
8. Neurologic disorder 8. Neurologic disorder
9. Hematologic disorder 9. Hemolytic anemia
10. Immunologic disorder (positive anti-dsDNA 10. Leukopenia OR lymphopenia
antibody OR positive anti-Sm antibody OR positive
antiphospholipid antibody (includes presence of a
lupus anticoagulant, false-positive treponemal
test, positive anticardiolipin antibody)
11. Positive ANA 11. Thrombocytopenia

ANA, Antinuclear antibody.

expression of B cell trophic factors). The nature of antigen presentation


TABLE 26.1 Frequency of Kidney
also may be important, with certain nuclear antigens capable of trig-
Manifestations in Lupus Patients With Renal gering an immunogenic response through interactions with a variety
Involvement of intracellular Toll-like receptors.
Manifestation Prevalence (%)
Proteinuria 100 PATHOGENESIS OF LUPUS NEPHRITIS
Nephrotic syndrome 45-65
Autoantibodies are crucial to the pathogenesis of LN, and the hallmark
Hematuria
of LN is the accumulation of immune complexes in glomeruli.19 Patients
Microscopic 80 with LN have autoantibodies against dsDNA, Sm antigen, C1q, nucleo-
Gross 1-2 somes, and other antigens. There is direct binding of dsDNA antibodies
Red blood cell casts 10 to the glomerular basement membrane (GBM), and cross-linking of
Cellular casts 30 positively charged nucleosome components such as chromatin between
Reduced renal function 40-80 autoantibodies and GBM.19 In proliferative LN, immune complexes are
Rapidly progressive 10-20 found in the subendothelial space, whereas in membranous LN, immune
glomerulonephritis complexes are found in the subepithelial space. The localization of
Acute kidney injury 1-2 immune complexes within glomeruli is influenced by size, charge, speci-
Hypertension 15-50 ficity, and avidity of the antigen and antibody, as well as by the clearance
Hyperkalemia 15 capacity of mesangium and local hemodynamics.
Glomerular immune complexes, especially in the subendothelial
space, activate proinflammatory mechanisms, including the complement
pathway, leukocyte Fc receptors, cytokines that regulate cell prolifera-
mimicry, in which there is exposure to viral or bacterial peptides with tion, and matrix formation and procoagulant factors.19-21 Nucleosomes
sequences similar to those of native antigens, also may lead to induction also can activate resident dendritic cells through binding to Toll-like
of autoantibody-producing cell lines. Both autoreactive B and T cells receptors 2 and 9.22 These activated pathways result in complement-
clonally expand in lupus, facilitated by failure of apoptotic mechanisms mediated kidney damage, intraglomerular hypertension and coagulation,
to silence autoreactive cells (i.e., loss of tolerance as well as an increased and Fc receptor–mediated leukocyte infiltration with release of proteolytic
308 SECTION IV Glomerular Disease

enzymes. Subepithelial immune complexes are associated with less can develop silently as a result of multiple pulmonary emboli or intra-
inflammation but increased production of GBM components and more vascular coagulation in association with antiphospholipid antibodies,
podocyte injury. or may be caused by nonthrombotic pulmonary arterial disease. Mitral
T cells contribute to the progression of LN by facilitating B cell dif- valve prolapse, and less commonly Libman-Sacks endocarditis, are both
ferentiation and expansion. Additionally, T helper cell (Th1) cytokines seen in SLE. Splenomegaly and lymphadenopathy are present in about
are overexpressed in kidneys of patients with LN and promote intrarenal one fourth of patients, and hematologic abnormalities can affect all three
inflammation by activation of macrophages, complement, and the Fc cell lines. SLE-associated anemia can result from impaired erythropoiesis,
receptor pathway. Kidney biopsies of LN patients also contain Th17 autoimmune hemolysis, or bleeding. Thrombocytopenia and leukopenia
cells, which secrete interleukin-17 (IL-17), thought to sustain renal may occur as part of the disease process or as complications of therapy.
inflammation by driving T cells away from maturing into a regula- Thrombotic events are not uncommon and should prompt a search for
tory phenotype (CD4+CD25hiFoxP3+) capable of suppressing autoan- antiphospholipid antibodies and other pro-coagulant abnormalities.
tibody production and attenuating the immune response.23 CD8 T cells
may gradually lose effector function and express inhibitory receptors
under persistent antigen exposure, and in essence become “exhausted.”
DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS
Although exhaustion may, for example, result in an inability to clear Although the diagnosis of lupus may be obvious in a young woman
a viral infection, it may be protective against relapse in autoimmune with classic manifestations and serologic markers, less typical presenta-
diseases. LN patients whose peripheral T cells displayed an exhausted tions are common and often result in multiple physician consultations
phenotype had a nonrelapsing disease course.24 It is unknown whether and diagnostic delay. This is in part a result of the varied features of
T cell exhaustion can be translated into a therapeutic approach for the disease and because signs and symptoms evolve over time. The
SLE. presence of four or more ACR criteria carries a 96% sensitivity and
In SLE and LN, neutrophils undergo a novel form of cell death specificity for lupus (see Box 26.1). However, the ACR diagnostic criteria
called NETosis, in which a chromatin meshwork (or NET) is released.22 were developed for clinical studies and do not always prove useful in
These NETs are a source of autoantigens and are not properly degraded an individual patient. In the SLICC classification system (see Box 26.1),
in lupus patients. In addition, lupus patients have an increased number SLE can be diagnosed with biopsy-proven immune complex nephritis
of low-density granulocytes that are more susceptible to NETosis. NET in the presence of ANA or anti-dsDNA, without other extrarenal disease
material has been found in LN biopsy samples, and the subset of SLE manifestations.
patients with LN often do not degrade. The NETs induce production Several autoimmune diseases mimic the extrarenal manifestations
of IFN-α by plasmacytoid dendritic cells, which are also found in LN of SLE, and some are associated with kidney involvement and GN,
kidneys.22 including fibromyalgia, Sjögren syndrome, thrombotic microangiopa-
Within the tubulointerstitial compartment, T and B cells are often thies, primary antiphospholipid syndrome, dermatomyositis, systemic
found in close proximity to each other and appear to be interacting, sclerosis, and mixed connective tissue disease. Conversely, several common
in some cases even forming germinal centers.25 Interstitial B cells aggre- forms of GN must be distinguished from LN because of similar clinical
gating with T cells may show clonal expansion and somatic hypermuta- features, including immunoglobulin A (IgA) vasculitis (Henoch-Schönlein
tion, suggesting the possibility of intrarenal autoantibody production purpura, see Chapter 23), antineutrophil cytoplasmic antibody (ANCA)–
against kidney-specific antigens, such as vimentin. Such interactions associated GN (see Chapter 25), bacterial endocarditis, and cryoglobu-
may contribute to interstitial inflammation in LN, and interstitial injury linemia. ANCAs of uncertain significance are detected in 20% of LN
is a major determinant of long-term renal survival.25 patients.

Immunologic Tests in Lupus


CLINICAL MANIFESTATIONS Antinuclear antibodies (ANAs) are found in more than 90% of untreated
Immune complex kidney involvement (LN) is usually heralded by patients with lupus. Although highly sensitive, neither the presence nor
proteinuria and active urinary sediment with microhematuria, dys- pattern (e.g., diffuse, speckled) are specific for SLE. Autoantibodies
morphic urine erythrocytes, and erythrocyte casts (see Table 26.1). against dsDNA are more specific, being present in 75% of untreated
More severe LN may present with the nephritic syndrome in associa- lupus patients, but are less sensitive than ANAs. Whereas high titers of
tion with proliferative GN, hypertension, and a decline in glomerular anti-dsDNA antibodies correlate with the presence of SLE and are often
filtration rate (GFR). Less frequently, renal disease in lupus presents used to follow the course of LN, antibodies to single-stranded DNA
as a tubulointerstitial disorder such as renal tubular acidosis (Chapter (ssDNA) are found in many rheumatologic conditions and do not cor-
12), isolated interstitial nephritis, or as a thrombotic microangiopathy relate with the course of LN. Sm antibodies are strongly associated with
associated with or without a antiphospholipid antibody syndrome (see the diagnosis of lupus and the presence of nephritis but are present in
Chapter 28). only about 25% to 30% of patients. Antibodies to C1q (anti-C1q) have
been more closely associated with the activity of LN than anti-dsDNA
Extrarenal Manifestations antibodies and may have a prognostic role in the follow-up of patients
Patients with active SLE often present with nonspecific complaints of with LN.26
malaise, low-grade fever, poor appetite, and weight loss. Other common Serum levels of total hemolytic complement and complement com-
features include patchy alopecia, oral or nasal ulcerations, arthralgias, ponents C3 and C4 are often depressed in untreated SLE and especially
nondeforming arthritis, and a variety of skin findings, including pho- in LN. In general, both C3 and C4 are depressed. Preferential C4 depres-
tosensitivity, Raynaud’s phenomenon, and a “butterfly” (malar) facial sion in lupus patients may reflect activation of the classic complement
rash. Livedo reticularis is seen in up to 15% of cases and may be associ- pathway. In some cases, low C4 with normal C3 in a patient with lupus
ated with miscarriage, thrombocytopenia, and antiphospholipid anti- may reflect genetic C4 deficiency or the presence of cryoglobulins.
bodies. Neuropsychiatric involvement presents with headache, nerve Preferential depression of C3 is observed in patients with postinfectious
palsies, psychoses, or frank coma. Serositis, in the form of pleuritis glomerulonephritis (PIGN) and the C3 glomerulopathies (see Chapters
or pericarditis, affects up to 40% of patients. Pulmonary hypertension 22 and 55) rather than in SLE/LN.
CHAPTER 26 Lupus Nephritis 309

TABLE 26.2 The ISN/RPS Classification of Lupus Nephritis and Associated Clinical
Correlations
Class Definition Urine Findings Clinical Findings
I: Minimal mesangial LN Normal glomeruli by LM, but Usually unremarkable None relevant to kidney;
mesangial immune deposits by IF excellent renal prognosis*
II: Mesangial proliferative LN Mesangial hypercellularity and/or Microscopic hematuria; Preserved renal function;
expansion with mesangial proteinuria, if present, hypertension infrequent;
immune deposits is usually low-grade excellent renal prognosis*
III: Focal LN Segmental or global endocapillary Microscopic Hypertension possible; renal
III (A): Purely active lesions: focal proliferative LN or extracapillary hematuria; insufficiency and nephrotic
III (A/C): Active and chronic lesions: focal glomerulonephritis affecting less proteinuria syndrome not unusual;
proliferative and sclerosing LN than 50% of glomeruli with variable renal prognosis
III (C): Chronic inactive lesions with glomerular mesangial and subendothelial
scars: focal sclerosing LN immune deposits
IV: Diffuse LN Segmental (S) or global (G) Microscopic Hypertension; renal
IV-S (A) or IV-G (A): Purely active lesions: diffuse endocapillary or extracapillary hematuria; insufficiency and nephrotic
segmental (S) or global (G) proliferative LN glomerulonephritis affecting 50% proteinuria syndrome frequent; variable
IV-S (A/C) or IV-G (A/C): Active and chronic or more of glomeruli with renal prognosis
lesions: diffuse segmental or global mesangial and subendothelial
proliferative and sclerosing LN immune deposits
IV-S (C) or IV-G (C): Inactive with glomerular scars:
diffuse segmental or global sclerosing LN
S: >50% of affected glomeruli have segmental lesions
G: >50% of affected glomeruli have global lesions
V: Membranous LN Glomerular basement membrane High-grade proteinuria; Preserved renal function;
thickening with subepithelial and microscopic nephrotic syndrome common;
mesangial immune deposits hematuria possible renal prognosis good*
Anti-PLA2R antibody negative
VI: Advanced sclerosing LN ≥90% of glomeruli globally Microscopic hematuria; Renal insufficiency/failure
sclerosed without residual proteinuria not expected
disease activity unusual

IF, Immunofluoresence microscopy; LM, light microscopy; LN, lupus nephritis.


*As long as no transformation to a proliferative class.

RENAL BIOPSY IN SYSTEMIC


LUPUS ERYTHEMATOSUS
Although nephritis may be suspected based on clinical symptoms and
laboratory markers, a renal biopsy is required for confirmation, subclas-
sification, prognosis, and management decisions. Indications for biopsy
include persistent proteinuria greater than 0.5 mg/mg (0.5 g/day), glo-
merular hematuria or leukocyturia without proteinuria, and/or unex-
plained fall in estimated GFR (eGFR). The risks for biopsy-related bleeding
are increased when serologic features of the antiphospholipid syndrome
are present. A repeat renal biopsy can be considered when there is dete-
rioration or persistence in markers of renal disease with uncertainty as
to future treatment.
Fig. 26.1 ISN/RPS class I: minimal mesangial lupus nephritis.
PATHOLOGY Light microscopy is normal, but immunoperoxidase shows C1q localiza-
tion (associated with IgG and C3) throughout the mesangial area (mag-
Although LN may affect all the structures of the kidney, glomerular nification 400X).
involvement has been the best characterized. Often, the degree and type
of glomerular involvement correlates with the clinical presentation and Less common glomerular pathologies omitted from the ISN/RPS system
disease course and also influences treatment decisions. LN is currently include lupus podocytopathy (see later discussion) and pauci-immune
classified by the International Society of Nephrology (ISN)/Renal Pathol- crescentic GN (analogous to ANCA-associated renal vasculitis).
ogy Society (RPS) system (Table 26.2), which is based on glomerular Although IgG is generally the dominant glomerular immunoglobulin
histology using light and immunofluorescence microscopy. Examples in LN, IgA and IgM along with the complement components C1q and
of the different histologic classes are shown in Figs. 26.1 through 26.5. C3 are often seen as well, giving the “full-house” pattern that is highly
310 SECTION IV Glomerular Disease

B
Fig. 26.2 ISN/RPS class II: lupus nephritis (mesangial disease).
(A) Mesangial expansion but little increase in tuft cellularity (magnifica-
tion 400X), and the peripheral capillary walls are normal. (Silver meth-
enamine stain.) (B) Extensive mesangial IgG deposits shown by
immunoperoxidase (magnification 400X); the aggregates are just beginning
to invade peripheral capillary walls.

suggestive of LN. Strong glomerular C1q staining is also suggestive of


LN. Fibrin staining, corresponding to active lesions, is often noted in
C
the glomerular tuft, especially in crescents, if present.
On electron microscopy (EM), the distribution of immune deposits Fig. 26.3 ISN/RPS class III: focal proliferative lupus nephritis.
corresponds to immunoglobulin deposits identified by immunohistol- (A) Low-power magnification shows focal and segmental proliferative lesion–
ogy. Some electron-dense immune deposits have an organized substruc- active (class IIIA) with less than 50% of glomeruli affected (magnification
ture known as fingerprinting, corresponding to the presence of curvilinear 100X). (Hematoxylin-eosin [HE] stain.) (B) Area of focal necrosis containing
microtubular or fibrillar structures in the deposits. Tubuloreticular cellular debris, karyorrhexis (arrow), is surrounded by an area of cellular
inclusions, which are tubular structures located in the endoplasmic proliferation (magnification 400X). (Silver methenamine/HE.) (C) Major focal
and segmental proliferative lesion is affecting almost half the glomerular
reticulum of renal endothelial cells, are often found in biopsy specimens
capillary tuft (magnification 400X). (Hematoxylin/lissamine green.)
of LN patients and are thought to reflect a high interferon milieu.

Tubulointerstitial and Vascular Disease True vasculitis is rare in LN, but several other vascular lesions can
A deficit of the ISN/RPS classification system is that it does not be seen (Fig. 26.7). For example, fibrinoid vessel necrosis or thrombotic
assess the tubulointerstitial or vascular compartments of the kidney, microangiopathy may be found in severe proliferative LN.
even though interstitial and vascular damage predicts kidney out-
comes.27 About 50% of patients with LN, predominantly those with Transformation of Histologic Appearance and
proliferative glomerular lesions, have immune aggregates along tubular “Silent” Lupus Nephritis
basement membranes. Interstitial inflammatory cell infiltrates, includ- The ISN/RPS class found at diagnostic kidney biopsy for LN is not
ing T cells, B cells, and monocytes are frequently found, and tubulitis fixed for an individual’s entire clinical course. Those successfully
can be seen in active disease (Fig. 26.6). In chronic disease, the inter- treated for proliferative (class III/IV) LN may transform to a less
stitium is often expanded by fibrosis and sparser infiltrates. Infre- serious histology, such as class II, or have resolution of inflammatory
quently, tubulointerstitial nephritis is seen in the absence of lesions with scarring and move from an active to a chronic histologic
glomerular disease and may result in acute kidney injury or renal pattern. Conversely, patients initially diagnosed with class II or V
tubular acidosis. LN may transform to proliferative LN, usually class IVG (global).
CHAPTER 26 Lupus Nephritis 311

Clinical, Laboratory, and Histopathologic Correlations


and Outcomes
Given the highly variable clinical presentations of LN and the overlap
in the clinical features of patients with different types of glomerular
injury, kidney biopsy remains the gold standard for diagnosis and sub-
sequent management of LN. Table 26.2 summarizes the typical clini-
copathologic correlations of LN. Lupus membranous nephropathy may
be distinguished from idiopathic membranous nephropathy by the
absence of anti-PLA2r autoantibodies in the circulation and by the
presence of all IgG isotypes in the immune deposits, although restric-
tion to the IgG4 isotype has been rarely observed in LN patients.
The importance of the kidney biopsy is underscored by the significant
A number of patients with SLE and kidney injury who do not have classic
LN. For example, up to 24% of patients may have a renal thrombotic
microangiopathy, either alone or associated with LN.30 A small (1.3%)
but important subset of patients with SLE present with nephrotic syn-
drome, with or without acute kidney injury, but their kidney biopsies
appear normal on light microscopy, mimicking minimal change disease
(MCD), or show segmental sclerosis, mimicking focal segmental glo-
merulosclerosis (FSGS).31 EM shows diffuse podocyte effacement, without
immune complexes. These patients, although not classified by ISN/RPS,
have lupus podocytopathy and like MCD or FSGS, often respond to
corticosteroids alone. A recent Chinese study showed that remission in
response to corticosteroids for 12 weeks occurred in 75% of patients
with podocytopathy and more than 90% when those with FSGS-like
lesions were excluded.32 The risk for relapse exceeded 50%, and at repeat
kidney biopsy half of the relapsed patients had transformed to class IV
B or V LN. Although these results may not be generalizable to a non-Asian
population, they underscore the importance of histology-driven treat-
ment decision analysis.

Long-Term Prognosis and Kidney Histology


Features of reversible (active) or irreversible (chronic) damage on
kidney biopsy are captured by the National Institutes of Health (NIH)
activity and chronicity indices (Table 26.3). Similar to other types of
GN, long-term kidney prognosis is poor if the biopsy sample shows
extensive glomerulosclerosis or interstitial fibrosis and tubular atrophy.
In patients with less extensive scarring the activity and chronicity indices
at diagnostic biopsy may not correlate with the course of LN.28 In
contrast, after treatment, persistent activity (activity index >2) on repeat
kidney biopsy is associated with a worse long-term renal prognosis;
C high chronicity (chronicity index >6) at repeat biopsy shows a similar
trend.28
Fig. 26.4 ISN/RPS class IV: lupus nephritis. (A) Active, diffuse proliferative
lupus nephritis (magnification 100X). (B) Immunoperoxidase staining shows dense,
irregular aggregates of IgG along the peripheral capillary walls (magnification TREATMENT
400X). (C) Electron microscopy reveals the immune aggregates as electron-dense
deposits (arrows), predominantly in the subendothelial location. It is useful to divide the treatment of patients with active proliferative
LN into initial and maintenance phases. The initial or induction phase
addresses acute life- or organ-threatening disease. The maintenance
This class switch is often heralded by increasing proteinuria and phase focuses on consolidating remissions, preventing relapses and the
activity of the urine sediment.28 Switching between class IVS (seg- long-term management of chronic, more indolent disease.
mental) and G subtypes is not seen and may reflect different patho- The ISN/RPS biopsy classification should guide initial therapy (see
genic mechanisms. Table 26.2). In general, the immunosuppressive treatment of extrarenal
The concept of silent LN has been proposed based on kidney biopsies lupus manifestations is sufficient for class I and II LN. The combination
done in patients with SLE but no clinically obvious kidney involve- of high-dose corticosteroids plus an immunosuppressive agent is mainly
ment.29 A few hundred such patients have been reported, with normal used for patients with active focal proliferative LN (classes IIIA and
kidney function and no hematuria or proteinuria but with immune IIIA/C), active diffuse proliferative LN (classes IVA and IVA/C), and
complexes in their glomeruli and a histologic pattern most often con- membranous lupus (class V).
sistent with class I/II LN. A few such patients even display class III, IV, Although the goal of initial therapy is to induce a complete renal
or V LN. Silent LN may represent a preclinical stage in the evolution remission, this occurs in only 30% to 40% of LN patients by 12 months.33
of LN. Most studies define complete renal response as a reduction in proteinuria
312 SECTION IV Glomerular Disease

US D D
S

BM

A B C
Fig. 26.5 ISN/RPS class V: membranous lupus. (A) Thick (~0.5 mm) araldite-embedded section stained
with toluidine blue shows not only the extramembranous material in dark blue (arrow) but also the presence
of mesangial deposits, which are common in lupus membranous nephropathy (magnification 400X). (B) Silver
methenamine–stained section shows some double contouring of the silver-positive basement membrane
(arrow) and subendothelium-deposited material, as well as the characteristic silver-positive spikes of base-
ment membrane–like material (magnification 400X). (C) Electron micrograph shows the predominantly sub-
epithelial electron-dense deposits (D) separated by protrusions of basement membrane material (spikes, S).
BM, Basement membrane; US, urinary space.

A B
Fig. 26.6 Interstitial lupus nephritis. (A) Interstitial infiltrate invading and destroying tubules (tubulitis).
Tubular basement membranes, which stain black with silver, are digested in the areas of tubulitis (arrow)
(magnification 400X). (B) Immunofluorescence shows aggregates of C3 in the tubular basement membrane
(right) as well as within the glomerulus (left). Such tubular basement membrane aggregates are common in
lupus nephritis, being found in 60% to 65% of biopsy specimens overall and with increasing frequency from
class II (20%) to class IV (75%).

TABLE 26.3 The Point System Used to Calculate Lupus Nephritis Biopsy Activity and
Chronicity Indices*
LESIONS CONTRIBUTING TO
LESIONS CONTRIBUTING TO ACTIVITY INDEX CHRONICITY INDEX

Large
Semiquan- Subendo-
titative Glomerular thelial Interstitial
Lesion Cellular Necrosis: Glomerular Endocapillary Immune Inflamma- Glomerular Fibrous Tubular Interstitial
Score* Crescents Karyorrhexis Neutrophils Proliferation Deposits tion Sclerosis Crescent Atrophy Fibrosis
None 0 0 0 0 0 0 0 0 0 0
Mild 2 2 1 1 1 1 1 1 1 1
Moderate 4 4 2 2 2 2 2 2 2 2
Severe 6 6 3 3 3 3 3 3 3 3

*Maximum activity index is 24; maximum chronicity index is 12.


CHAPTER 26 Lupus Nephritis 313

therapy in a White population with mild-moderate disease showed


comparable complete and partial remission rates at 2 years.36 An obser-
vational study (RITUXILUP) used anti–B cell therapy (rituximab) plus
2 doses of intravenous methylprednisolone followed by maintenance
with mycophenolate mofetil (MMF) in the absence of oral steroids.37
The overall complete plus partial renal response rate was 88%, with a
median time to remission of 9 months. During the follow-up period
19 LN flares occurred at a median of 24 months after remission and
79% were managed without oral steroids.

Immunosuppressive Agents
Although corticosteroids effectively control proliferative LN, long-term
kidney function was better preserved with fewer LN relapses at 3 to 5
years if corticosteroids were combined with cyclophosphamide during
Fig. 26.7 Vascular damage in lupus nephritis. Thrombus (arrow)
initial therapy.38 These data highlight the need for long-term follow-up
occludes a glomerular capillary loop in this class IV biopsy specimen. in assessing initial LN therapies. Both daily oral and intravenous pulses
Such a thrombus contains platelets and cross-linked fibrin as well as of cyclophosphamide are effective in LN, although intravenous cyclo-
immunoglobulins and thus has some characteristics of true thrombus phosphamide given as 6-monthly pulses of 0.5 to 1 g/m2 (NIH protocol)
(magnification 400X). (Silver methenamine/hematoxylin stain.) has been the standard of care for several years. The Euro-Lupus Nephritis
Trial compared lower dose (500 mg) cyclophosphamide intravenously
every 2 weeks for 3 months (total of 3 g) followed by azathioprine
to less than 0.5 or a ratio of urine protein to creatinine of less than maintenance to the NIH protocol, and similar efficacy with less toxicity
0.5 absence of glomerular hematuria or red blood cell (RBC) casts, and fewer infections short term39 and after 5 to 10 years.40 Although
and normalization or stabilization of GFR. The predictive value of the Euro-Lupus cyclophosphamide regimen was initially tested in a
glomerular hematuria in the definition of remission has recently been mainly White population, similar rates of remission induction are
questioned.34 It may be best to view induction as the first step of con- achieved with low-dose cyclophosphamide in Black, Hispanic, and
trolling renal inflammation to allow healing of kidney injury, which Southeast Asian patients.41,42
translates clinically into stabilization or improvement of renal function, Several randomized clinical trials involving multiethnic cohorts have
attenuation of proteinuria, and reduction in urine sediment activity. shown that oral MMF plus corticosteroids for 6 months followed by
However, all of these kidney parameters may not normalize by the end maintenance therapy is at least as effective as 6-monthly pulses of cyclo-
of induction. The term partial response has required a 50% reduction phosphamide (NIH protocol) plus corticosteroids followed by main-
in proteinuria to subnephrotic levels and stability or improvement in tenance therapy.33 MMF also showed similar treatment response rates
GFR and is usually achieved before complete response criteria are met. to the Euro-Lupus regimen at 24 weeks.42 Cyclophosphamide has been
Evidence in support of specific therapeutic agents and regimens favored in patients presenting with marked renal impairment or severe
should be treated with caution because of small study sizes, uncontrolled class IV LN on biopsy, but there are no convincing differences in outcome
or nonblinded methods, and heterogeneity in both LN presentations between MMF and cyclophosphamide induction for such patients.43
and responses to therapy. Therapeutic toxicity is a major contributing No improvement in overall mortality or severe infections has been seen
factor to morbidity and mortality, so claims of efficacy must be bal- with MMF; gastrointestinal disturbances are more frequent, but the
anced against adverse events. Other factors, such as ethnicity, referral risk for amenorrhea is lower than with cyclophosphamide.
practice, center experience, and use of concomitant medications vary Optimal MMF dosing and adjustment for different patient subgroups
across studies and over time, reducing the generalizability of some remains unclear. A dosing range of 2 to 3 g/day has been recommended
study results. as a target with dose reduction for intolerance and in certain ethnic
In addition to the immunosuppressive regimens discussed next, the subgroups. The MMF metabolite mycophenolic acid (MPA) has been
kidney protective measures outlined in Chapter 79 should be used as pharmacokinetically monitored as the area under the concentration-
appropriate. Furthermore, unless contraindicated, all patients with LN time curve (AUC) in patients receiving MMF for LN induction or
should be treated with an antimalarial agent because this was associated maintenance. However, monitoring yields highly variable AUCs after
with a lower risk for developing LN and ESRD and improved responses empiric MMF dosing with no correlation between AUCs and trough
to LN treatment.35 drug levels, and as yet there is no consensus on a target MPA AUC and
no correlation of adverse events with AUC.44
Proliferative Lupus Nephritis: Induction Long-term follow-up of patients who participated in clinical trials
Corticosteroids of MMF versus cyclophosphamide found no differences in renal outcome,
High-dose corticosteroids are used in all current induction regimens although one study reported a non-significant increase in renal flares
and are considered the standard of care. Prednisone (or prednisolone) is in patients induced with MMF.45 Differences in response between ethnic
most often started at 0.5 to 1 mg/kg/day ideal body weight (no more than and geographical subgroups have been suggested but not confirmed.46
80 mg/day) and then reduced to approximately 10 mg/day or less by 3 to However, a retrospective analysis of a Korean cohort showed similar
6 months. There is variation in the use of intravenous methylprednisolone remission rates for MMF and cyclophosphamide, but more relapses
infusions (0.5 to 1 g daily for 1 to 3 days) either as a routine component and a higher incidence of ESRD in the MMF group.47 A meta-analysis
of induction therapy followed by lower oral corticosteroid dosing or for showed a reduction in the 10- and 15-year risk of ESRD from LN
nephritis perceived to be severe. Adverse effects of corticosteroids have between 1970 and the mid-1990s, coincident with the use of cyclo-
led to attempts to minimize prolonged courses of high-dose corticoste- phosphamide as standard-of-care induction therapy. It is of some concern
roid therapy in lupus patients. A study comparing cyclophosphamide that from the mid-1990s until the late 2000s ESRD plateaued, but then
alone to cyclophosphamide plus high-dose corticosteroids as initial increased slightly, coincident with the era of MMF induction.4
314 SECTION IV Glomerular Disease

Treatment of Proliferative Lupus Nephritis

Class IIIA, IIIA/C, IVA, IVA/C  Class VInitial (Induction) Phase

Severe proliferative LN: Give intravenous methylprednisolone 0.5-1 g/d for 1-3 days before oral
Oral predniso(lo)ne 1 mg/kg/d ideal body weight, maximum 80 mg/d, taper over several
weeks
PLUS:

Intravenous Oral cyclophosphamide Low-dose intravenous


Oral MMF 2-
cyclophosphamide 1-1.5 mg/kg/d, cyclophosphamide
Or 3 g/d for 6 Or Or
0.5-1 g/m2 monthly maximum 150 mg/d 500 mg every 2 weeks for
months
for 6 months for 2-4 months 3 months

Maintenance phase*
predniso(lo)ne 5-10 mg/d

PLUS: MMF 1-2 g/d (first choice)


-or-
AZA 1-2.5 mg/kg/d (individual indication)
-or-
Cyclosporine 2.5-4 mg/kg/d (if MMF/AZA not tolerated)
*Assumes no worsening -or-
of LN and preferably a Tacrolimus to trough of 4-6 ng/ml (if MMF/AZA not tolerated)
response to initial therapy

Fig. 26.8 Approach to the induction and maintenance treatment of proliferative lupus nephritis
(LN). AZA, Azathioprine; MMF, mycophenolate mofetil.

The ACR, the Kidney Disease: Improving Global Outcomes (KDIGO), withdrew because of adverse events at 6 months, despite lower rates of
and the European League Against Rheumatology/European Renal Asso- leukopenia and upper gastrointestinal symptoms in this group.53 Although
ciation (EULAR/ERA) have independently developed evidence-based short-term, a preliminary report indicated a higher 6-month complete
guidelines for LN therapy. These are synthesized in the treatment algo- renal response rate (33% vs. 19%) in a multiethnic LN cohort treated
rithm presented in Fig. 26.8.48-50 with the CNI voclosporin plus corticosteroids and MMF compared
with corticosteroids and MMF alone.54 In future studies, it will be
Other Immunosuppressive Strategies important to verify preservation of long-term kidney function and
Azathioprine (AZA) has been used in combination with corticosteroids improvements in histology in CNI-treated LN patients. Furthermore,
for the induction of remission in proliferative LN. A randomized trial it is not clear that proteinuria is an appropriate renal remission end-
comparing AZA with cyclophosphamide found no difference in eventual point for comparing a CNI-based regimen to other drugs because of
outcome but more relapses, doubling of serum creatinine concentration, the known nonimmune antiproteinuric effects of CNIs.
and more chronicity on repeat biopsy in the AZA group.51 Although
AZA is not recommended as first-line therapy by the major LN guide- Biologic Agents
lines, it remains an option when MMF or cyclophosphamide are unavail- Rituximab, an anti-CD20 monoclonal antibody that depletes B cells, has
able, undesirable, or contraindicated. led to improved disease control in patients with LN with relapsing or
The calcineurin inhibitors (CNIs) tacrolimus and cyclosporine have refractory disease in retrospective and nonrandomized trials. However,
been tested as LN induction therapies and compared favorably in the in a large randomized, prospective trial, when used in combination with
short term.33 A trial from Hong Kong randomized 150 patients to tacro- MMF and corticosteroids for induction, rituximab did no better at 1
limus or MMF for 6 months, with responders receiving AZA for main- year than placebo.55 The role of rituximab in LN is therefore currently
tenance.52 Complete remission rates were approximately 60% in each uncertain, but it may be considered when other therapies have failed.
group, although there was a trend toward more renal flares at 5-year Abatacept, a fusion protein of CTLA4 and the immunoglobulin
follow-up in the tacrolimus group. Additionally, the primary proteinuria heavy chain, blocks T and B cell co-stimulation. In two large random-
end-point was less than 1 g/day, which may have inflated the complete ized controlled trials of proliferative LN, abatacept proved no better
response rate. Another Asian study took a multitargeted approach, than placebo on a background of MMF and corticosteroids56 or a back-
randomizing patients to tacrolimus plus MMF and corticosteroids or ground of low-dose cyclophosphamide and corticosteroids.41 However,
the NIH protocol of cyclophosphamide plus corticosteroids. The mul- a retrospective analysis of data from the first trial using different defini-
titarget induction regimen was associated with a higher remission rate tions of remission suggested that abatacept may have been effective
than cyclophosphamide but more patients in the multitarget group with somewhat less stringent response criteria.
CHAPTER 26 Lupus Nephritis 315

Treatment of Resistant Severe Proliferative Lupus Nephritis

LN treated with an Switch to an


MMF or CYC-based alternative initial
initial regimen No Improvement by regimen
3-6 months or
worsening at any time
No Improvement by
3-6 months or
worsening at any time

Consider LN as
Refractory

Rituximab 1g Cyclosporine 2.5 mg/kg/d Other Options:


Repeat in 2 weeks -or- • Intravenous IgG
Tacrolimus 0.1 mg/kg/d • Immunoablative
PLUS cyclophosphamide
Oral corticosteroids rescued by GCSF or stem
cell bone marrow
transplant
• Leflunomide
• Plasma exchange or
immunoadosorption
• Mesenchymal stem cells

Fig. 26.9 Treatment of resistant proliferative lupus nephritis (LN). CYC, Cyclophosphamide; GCSF,
granulocyte colony-stimulating factor; IgG, immunoglobulin G; MMF, mycophenolate mofetil.

Other pathogenic mediators currently being targeted in SLE or LN Renal Response to Initial Therapy
with monoclonal antibodies are the B cell–stimulating cytokine BAFF Achieving complete remission of LN predicts a good long-term outcome
and the type 1 interferon, IFN-α. Studies using an anti–IL-6 antibody with more than 90% 5-year patient and renal survival rates compared
and an anti-TWEAK antibody failed to show efficacy in proliferative with only 69% and 45% for the group not achieving remission.10 Partial
LN.57,58 remissions are also associated with improved outcomes. Complete and
partial renal responses to MMF or cyclophosphamide range from 18%
Resistant Proliferative Lupus Nephritis to 85% at 6 months and 32% to 85% at 12 months, and about half of
There is no uniformly accepted definition of resistant or refractory LN. treated LN patients achieve a complete or partial response by 1 year,
Up to one third of proteinuric nonresponders to induction therapy with an additional 25% by 2 years.66 Predictors of remission at the start
have inactive histologic findings on repeat biopsy. Fig. 26.9 outlines of treatment have traditionally included lower baseline serum creatinine
therapies that have been attempted for refractory disease. Most of these concentration, lower baseline urinary protein excretion, favorable renal
therapies were investigated in small, uncontrolled, and retrospective histologic class, lower chronicity index, stable GFR after 4 weeks of
studies. therapy, and White race.
Rituximab has had some success in patients with refractory disease, A post-hoc analysis of the Euro-Lupus low-dose cyclophosphamide
benefitting about 30% of nonresponders.59 In a registry, three different trial found an absolute level of proteinuria less than 0.8 g/day 1 year
rituximab regimens often in combination with other immunosuppres- after starting induction treatment was the single best predictor of good
sives were tried in 68 patients, and after 12 months 31% were in complete renal outcome after at least 7 years of follow-up.34 The other 12-month
renal remission.60 Although no benefit was seen with the addition of predictors examined were serum creatinine concentration below 1 mg/
plasma exchange to immunosuppressive therapy for remission induc- dl (88 µmol/l) and fewer than 5 urine RBCs per high-power field. In
tion in LN, the removal of circulating antibodies and other immune another study after induction with low-dose cyclophosphamide, an
reactants may be considered for patients with high serologic activity absolute level of proteinuria less than 0.7 g/day 1 year after starting
and progressive refractory nephritis or thrombotic microangiopathy.61 induction treatment again was the single best predictor of good renal
Intravenous immunoglobulin and CNIs have shown benefit in small outcome after at least 7 years of follow-up .67 However, the negative
series of patients with resistant LN.62,63 Infusion of allogeneic mesen- predictive value of proteinuria alone was low in both analyses, and
chymal stem cells derived from umbilical cord or bone marrow has many patients who did not achieve proteinuria less than 0.7 to 0.8 g/
been tested in a cohort of patients with refractory LN.64 At 12 months, day at 1 year still maintained good kidney function over time. Addition-
about 23% of patients had a complete renal response, but 40% of patients ally, most of these patients were White.
were considered to have treatment failures. Finally, for patients with
life-threatening resistant disease, small pilot studies have tested total Proliferative Lupus Nephritis: Maintenance Therapy
lymphoid irradiation and immunoablation by high-dose cyclophos- Because it generally takes months for proteinuria and serum creatinine
phamide and antithymocyte globulin, with or without autologous stem concentrations to decrease to baseline levels after beginning LN treat-
cell reconstitution. These approaches have led to sustained treatment- ment,68 one important aspect of maintenance immunosuppression is to
free remissions, but toxicity and treatment-related mortality needs to consolidate renal responses into complete and partial remissions using
be carefully weighed against potential benefit.65 drugs with a lower side effect profile than induction drugs. Additionally,
316 SECTION IV Glomerular Disease

maintenance therapy should ideally prevent renal flares and attenuate concerns over long-term nephrotoxicity, CNIs are best reserved for
or abrogate the development of chronic kidney disease. Current strate- patients who are unable to take MMF or AZA.
gies for LN maintenance therapy are presented in Fig. 26.8. The duration of maintenance therapy, and whether maintenance
Early investigations showed maintenance cyclophosphamide was can ever be stopped in patients with LN remain open questions. In
superior to corticosteroids alone.38,69 Subsequent work demonstrated general, LN patients are placed on maintenance immunosuppression
that less intense maintenance immunosuppression with AZA or MMF for several years. Withdrawal of maintenance therapy is the subject of
was as effective as maintenance cyclophosphamide and was associated an ongoing RCT (NCT01946880). Therapy reduction was examined
with improved patient morbidity and survival.70 retrospectively in European LN patients who had achieved complete
Corticosteroids, tapered from the initial treatment period, are gener- remission with normal kidney function, proteinuria of 0.5 g/day or
ally continued during the maintenance period, but at low dose (see Fig. less, inactive urine sediment, and no extrarenal SLE signs or symptoms.74
26.8). Both daily and alternate-day corticosteroid regimens have been The cohort started with 73 patients, 20 of whom experienced flare when
used, but there is no consensus as to the optimal duration of cortico- prednisone was tapered to a very low dose. Therapy was completely
steroid therapy. stopped in the other 52 patients, a median of 73 months after beginning
Two randomized clinical trials compared AZA 2 mg/kg/day and induction therapy. These patients were then followed for a median of
MMF 2 g/day for LN maintenance. In multiethnic patients who received 172 months, and 61% never relapsed. There were 15 cases of LN and
induction with cyclophosphamide or MMF, MMF maintenance therapy 5 nonrenal flares that occurred at a median of 37 months after stopping
was better than AZA at preventing renal flares, preserving renal function maintenance. Risk factors for LN flare after withdrawal of therapy in
over 3 years, and delaying progression to ESRD.45 In a study of mainly this cohort included shorter duration of overall treatment, decreased
European White patients (MAINTAIN Nephritis trial), AZA and MMF use of antimalarials, and lack of maintenance cytotoxic drug use.
were equally effective at preventing renal flares over 4 years after initial
treatment with low-dose cyclophosphamide.71 Although MMF appears Membranous Lupus Nephropathy
to be the maintenance drug of choice for most LN patients, therapy Membranous nephropathy is often diagnosed in association with pro-
should be individualized. In some patients, AZA may be preferred, such liferative forms of LN. In these patients, treatment is directed at the
as for patients in complete remission who want to become pregnant. proliferative component. Alternatively, the combination of low doses
Finally, two randomized trials compared the CNIs tacrolimus and of corticosteroids, MMF, and a CNI, the so-called multitarget regimen,
cyclosporine to AZA for maintenance.72,73 Although both trials were has shown considerable success in an Asian cohort of mixed membra-
underpowered and the tacrolimus trial had only 6 months of follow-up, nous and proliferative LN.75
both demonstrated that CNIs were as effective as AZA in preventing The optimal treatment of pure class V LN remains unclear, but
renal flares. Given fewer data for tacrolimus and cyclosporine, and a reasonable treatment algorithm is presented in Fig. 26.10. KDIGO

Treatment of Membranous Lupus Nephritis

Pure class V LN

Subnephrotic proteinuria Nephrotic proteinuria


-and- -or-
Normal renal function Abnormal renal function
-and- -and-
Asymptomatic Symptomatic

Renin-angiotensin- Renin-angiotensin-
aldosterone system aldosterone system
blockers blockers

Response Proteinuria MMF 2-3 g/d or AZA 1-2.5 mg/kg/d


remains 1 g/d  corticosteroids for 6 months

Intravenous cyclophosphamide 0.5-1 g/m2


monthly for 6 months
-or-
Cyclosporine 4-6 mg/d for 12 months No response Partial/Complete response
-or-
Tacrolimus 0.1-0.2 mg/kg/d for
No response: Choose an alternative from listed therapies
6-12 months
 corticosteroids
Maintenance with MMF 1-2 g/d or AZA
1-2.5 mg/kg/d

Fig. 26.10 Treatment of membranous lupus nephritis. AZA, Azathioprine; MMF, mycophenolate mofetil.
CHAPTER 26 Lupus Nephritis 317

guidelines suggest renoprotective and antiproteinuric therapies for disease, a frequent outcome of LN, is also a cardiovascular risk factor.
patients with subnephrotic proteinuria and no renal impairment, with Reduction of atherosclerotic risk should focus on blood pressure control
immunosuppression reserved for patients with nephrotic syndrome (goal of 130/80 mm Hg), use of statins and hydroxychloroquine to correct
and/or renal impairment.48 The only small RCT in patients with class lipid abnormalities,81 and reduction of inflammatory disease activity.
V LN compared cyclophosphamide or cyclosporine to corticosteroids The effects of SLE and LN on pregnancy and fetal outcomes, and
alone.76 The patients studied had preserved kidney function and a the effects of pregnancy on LN activity, are discussed in Chapter 43.
mean proteinuria of almost 6 g/day. Complete and partial remissions
were more frequent in the cyclophosphamide and cyclosporine-treated END-STAGE RENAL DISEASE AND RENAL
patients. Cyclosporine induced remission more rapidly than the other
drugs, but LN relapses were fewer in the cyclophosphamide group.
TRANSPLANTATION
Two trials of MMF versus cyclophosphamide as initial therapy of LN Although approximately 10% of all lupus patients develop ESRD, the
included 84 patients with pure membranous nephropathy among the overall proportion of patients with ESRD attributable to lupus is 1%
510 patients enrolled.77 Remissions, relapses, and courses were similar to 2%.3,82 Patients of African ancestry are at higher risk for ESRD. Extra-
in the class V patients treated with MMF and cyclophosphamide. renal lupus is often quiescent by the time patients reach ESRD, but
Azathioprine plus corticosteroids also has been successful in patients those with active extrarenal disease may require immunosuppression
with membranous LN, but these studies have been observational or while receiving renal replacement therapy. It has been suggested that
retrospective.78 patients with LN defer transplantation for 3 to 6 months to allow SLE
to become inactive; however, recent data suggest that there is an increased
Long-Term Monitoring of Lupus Nephritis Patients risk for allograft failure if LN patients wait more than 3 months before
The relapse (flare) rate for LN ranges from 35% to 60%, depending on transplantation.83 Interestingly, the correlation between longer wait times
the population studied, the criteria for relapse, and the maintenance and increased allograft failure was not seen in African American patients.
therapy used,78-80 suggesting that LN patients in remission be closely Survival of lupus patients on dialysis or after kidney transplantation is
followed. We recommend quarterly monitoring (including blood pres- similar to those of patients with other renal diseases.82 Thrombotic
sure, kidney function, proteinuria, urinary sediment, serum C3 and events are increased in LN recipients, especially if they are positive for
C4) with anti-dsDNA measured at least biannually.50 Although changes antiphospholipid antibody, but this does not appear to correlate to a
in serology alone do not warrant therapeutic action, they may serve as diminished allograft survival.82 Recurrent LN after kidney transplanta-
an early warning that autoimmune activity is increasing and patients tion is rare, occurring in only about 1% of patients.84
should be watched even more closely for flare.
Repeat kidney biopsies should be considered during long-term
management of LN.66 At LN flare, a repeat biopsy should be considered
REFERENCES
if a change in histologic class is suspected. Patients with proliferative 1. Petri M, Orbai AM, Alarcon GS, et al. Derivation and validation of the
LN tend to remain proliferative at flare, but patients with class II and Systemic Lupus International Collaborating Clinics classification criteria
V not infrequently develop a proliferative component. Evaluation of for systemic lupus erythematosus. Arthritis Rheum. 2012;64:2677–2686.
persistent proteinuria and declining kidney function in LN also may 2. Ines L, Silva C, Galindo M, et al. Classification of systemic lupus
benefit from a repeat biopsy. Several studies have shown considerable erythematosus: Systemic Lupus International Collaborating Clinics versus
discordance between histologic activity and clinical activity. Persistent American College of Rheumatology criteria. A comparative study of
2,055 patients from a real-life, international systemic lupus
proteinuria may be due to persistent inflammatory disease activity and
erythematosus cohort. Arthritis Care Res. 2015;67:1180–1185.
require further immunosuppression or may be due to chronic damage 3. Hanly JG, O’Keeffe AG, Su L, et al. The frequency and outcome of lupus
and nephron loss, in which immunosuppression will not be beneficial. nephritis: results from an international inception cohort study.
When considering reduction or withdrawal of maintenance therapy a Rheumatology. 2016;55:252–262.
repeat biopsy may help because even patients with sustained clinical 4. Tektonidou MG, Dasgupta A, Ward MM. Risk of end-stage renal disease
inactivity still may have histologic activity.66 in patients with lupus nephritis, 1971-2015: a systematic review and
Bayesian meta-analysis. Arthritis Rheum. 2016;68:1432–1441.
ANTIPHOSPHOLIPID ANTIBODY SYNDROME, 5. Bastian HM, Roseman JM, McGwin G Jr, et al. Systemic lupus
erythematosus in three ethnic groups. XII. Risk factors for lupus
ATHEROSCLEROSIS, AND PREGNANCY IN nephritis after diagnosis. Lupus. 2002;11:152–160.
LUPUS NEPHRITIS 6. Feldman CH, Hiraki LT, Liu J, et al. Epidemiology and
sociodemographics of systemic lupus erythematosus and lupus nephritis
Intrarenal thrombosis caused by the antiphospholipid antibody syn- among US adults with Medicaid coverage, 2000-2004. Arthritis Rheum.
drome is found in 30% of patients with SLE and may occur in the 2013;65:753–763.
presence or absence of LN (see Chapter 28). The mainstay of treatment 7. Yap DY, Tang CS, Ma MK, et al. Survival analysis and causes of mortality
for antiphospholipid nephropathy has been anticoagulation plus chlo- in patients with lupus nephritis. Nephrol Dial Transplant. 2012;27:
roquine or hydroxychloroquine, although immunosuppressive agents 3248–3254.
also have been used. 8. Lerang K, Gilboe IM, Steinar Thelle D, Gran JT. Mortality and years of
Patients with lupus have increased risk for atherosclerotic complica- potential life loss in systemic lupus erythematosus: a population-based
tions and greater atherosclerotic plaque burden compared with age- cohort study. Lupus. 2014;23:1546–1552.
matched controls, and these factors contribute to morbidity and mortality 9. Mok CC, Kwok RC, Yip PS. Effect of renal disease on the standardized
mortality ratio and life expectancy of patients with systemic lupus
in SLE.74 The risk for heart attack in a young woman with SLE is 50 times
erythematosus. Arthritis Rheum. 2013;65:2154–2160.
greater than that of a healthy woman, and even older women with SLE 10. Chen YE, Korbet SM, Katz RS, et al. Collaborative Study G., Value of a
have 2.5 to 4 times the risk for myocardial infarction of age-matched complete or partial remission in severe lupus nephritis. Clin J Am Soc
controls. After adjustment for all traditional cardiovascular risk factors, Nephrol. 2008;3:46–53.
patients with SLE have a 7- to 10-fold higher risk for nonfatal and a 11. Munroe ME, James JA. Genetics of lupus nephritis: clinical implications.
17-fold higher risk for fatal myocardial infarctions. Chronic kidney Semin Nephrol. 2015;35:396–409.

You might also like