Skip to main content
Clinical Journal of the American Society of Nephrology : CJASN logoLink to Clinical Journal of the American Society of Nephrology : CJASN
. 2018 Jul 24;13(8):1264–1275. doi: 10.2215/CJN.01920218

Complications of Immunosuppression in Glomerular Disease

J Ashley Jefferson 1,
PMCID: PMC6086710  PMID: 30042223

Abstract

Most glomerular diseases are immunologically mediated disorders of the kidney and are common causes of ESKD. In addition to supportive therapy, a wide range of immunosuppressive agents are used in the management of patients with these conditions. Immunosuppression requires a careful balance of risk and benefits, and many of these agents have a narrow therapeutic window and require close monitoring. This review describes the side effects of immunosuppressive agents used in recent randomized, controlled trials of glomerular disease, and highlights some of the key adverse events that determine the choice and prescription of these medications.

Keywords: glomerular disease; rituximab; glucocorticoids; mycophenolate mofetil; cyclophosphamide; tacrolimus; adverse events; lupus nephritis; ANCA; membranous nephropathy; IgA nephropathy; Immunosuppressive Agents; glomerulonephritis; immunosuppression; Kidney Failure, Chronic; kidney

Introduction

Immunosuppressive agents are used in the treatment of glomerular diseases, but are associated with a high risk of adverse effects. This review is intended as a guide for clinicians and focuses on the most commonly prescribed immunosuppressive agents, recognizing that we are in a time of rapid evolution with multiple new therapies being developed. Much of the recent evidence in glomerular disease is drawn from studies of lupus nephritis (LN) and ANCA-associated vasculitis (AAV) because in older studies of primary glomerular disease, the details of adverse events were often not well captured. It should be recognized that each of the medications described below has a wide range of adverse effects beyond the scope of this review, but are described in the package insert supplied with medications or may be reviewed in resources such as Micromedex.

Glucocorticoids

Glucocorticoids (GC) have both anti-inflammatory and immunosuppressive effects, and have a long list of side effects, including infection, bone disease, dysglycemia, obesity, hypertension, psychosis, gastrointestinal bleeding, cataracts, and long-term risks of cardiovascular disease.

Infection Risk

Specific infections that warrant consideration for screening and prophylaxis include Pneumocystis jirovecii pneumonia (PJP), tuberculosis, strongyloidiasis, hepatitis B (HBV) and hepatitis C (HCV), HIV, herpes zoster virus, and candidiasis. In a UK primary care database study (n=275,072), patients taking GC had a two to six fold increased rate of infections compared with those with the same underlying disease not exposed to steroids (1). Risk factors for infection included higher GC doses, increasing age, diabetes, and hypoalbuminemia. In observational studies in patients with rheumatology diseases, low-dose GCs (prednisone <10 mg/d) are associated with small increased risks of bacterial infections, but the risks of more severe opportunistic infections increase at higher doses (prednisone >20 mg/d) (2). Infection risk may be amplified in glomerular disease by urinary loss of Ig and complement, and by the immunodeficiency state of kidney disease.

In primary glomerular disease, steroid monotherapy is widely used in minimal change disease, FSGS, and IgA nephropathy. In three older studies of IgA nephropathy, severe adverse events (SAEs) due to infection were not described (35). By contrast, recent studies using high-dose steroids as monotherapy, including the Supportive Versus Immunosuppressive Therapy for the Treatment of Progressive IgA Nephropathy (STOP-IgAN) (6) and Therapeutic Evaluation of Steroids in IgA Nephropathy Global (TESTING) (7) trials (Tables 1 and 2), showed increased infection rates, partly because of more complete adverse event reporting. The TESTING trial was stopped early because of increased adverse events in the oral methylprednisone arm, mostly severe infections, including three patients with PJP (7). Notably, PJP prophylaxis was not used in this study, nor in other recent studies of IgA nephropathy. In a meta-analysis of early LN studies, the risk of major infection did not differ between GCs alone and GCs with the addition of azathioprine or cyclophosphamide (CYC) (8), highlighting the importance of steroid dose in the overall contribution to infection risk. In AAV, a large multicenter study, the Plasma exchange and glucocorticoid dosing in the treatment of anti-neutrophil cytoplasm antibody associated vasculitis (PEXIVAS), will soon report outcomes in patients treated with low- versus standard-dose GCs (Clinicaltrials.gov identifier NCT00987389).

Table 1.

Selected randomized controlled trials of IgA nephropathy

Study Study Design Efficacy Adverse Events
Rauen et al. (64) (STOP-IgA, IgA nephropathy) Prospective, multicenter, RCT (2008–2011); sample size: 162; intervention: immunosuppression (Pozzi protocol steroids or oral CYC [1.5 mg/kg 3 mo, then oral AZA 1.5 mg/kg 3 yr] versus supportive care only; end point: clinical remission, decline in GFR; follow-up: 3 yr No significant difference in the annual decline in eGFR or remission rates between the two groups Immunosuppression (n=82) Supportive care (n=80)
SAEs 29 (35%) 21 (26%)
Serious infections 8 (10%) 3 (4%)
Total infections 111 174
Cancer 2 (2%) 0
Diabetes/impaired glucose 9 (11%) 1 (1%)
Rauen et al. (6) (STOP-IgA, high GFR subgroup) Prospective, multicenter, RCT (2008–2011); sample size: 162; intervention: immunosuppression corticosteroids (Pozzi protocol) versus supportive care; end point: clinical remission, decline in GFR; follow-up: 3 yr Increased complete remission with steroid group (20% versus 3%) Steroids (n=54) Supportive care (n=55)
SAEs 12 (22%) 14 (25%)
Serious infections 4 (7%) 2 (4%)
Total infections 115 69
Cancer 0 0
Diabetes/impaired glucose 9 (17%) 1 (2%)
Lv et al. (7) TESTING, IgA nephropathy) Prospective, multicenter, RCT (2012–2015); sample size: 262; intervention: oral methylprednisone (0.6–0.8 mg/kg per d, taper after 2 mo) versus placebo; end point: ESKD or 40% decrease in GFR; follow-up: median 2.1 yr Study terminated early second to excess SAEs in oral methylprednisone group Oral methylprednisone (n=136) Placebo (n=126)
SAE 20 (15%) 4 (3%)
Serious infection 11 (8%) 0
Pneumocystis jirovecii 3 (2%) 0
Gastrointestinal SAE 4 (3%) 1 (1%)
Osteonecrosis 2 (1%) 0
Hou et al. (65) (IgA nephropathy) Prospective, multicenter, RCT (2010–2013); sample size: 176; intervention: MMF (1.5 g/d)+prednisone (0.4–0.6 mg/kg per d, taper) versus prednisone only (0.8–1.0 mg/kg per d, taper); end point: complete remission; follow-up: 12 mo No difference in complete remission rate (48% versus 53%) MMF+prednisone (n=87) High-dose prednisone (n=88)
SAE 5 (6%) 6 (7%)
Any infection 27 (31%) 20 (23%)
Serious infection 3 (3%) 4 (5%)
Diabetes (new) 1 (1%) 12 (14%)
Gastrointestinal 7 (8%) 10 (11%)
Osteonecrosis 0 1 (1%)

STOP-IgA, Supportive Versus Immunosuppressive Therapy for the Treatment of Progressive IgA Nephropathy; RCT, randomized controlled trial; CYC, cyclophosphamide; AZA, azathioprine; SAE, serious adverse event; TESTING, Therapeutic Evaluation of Steroids in IgA Nephropathy Global; MMF, mycophenolate mofetil.

Table 2.

Selected randomized controlled trials of primary membranous nephropathy

Study Study Design Efficacy Adverse Events
Praga et al. (66) (membranous nephropathy) Prospective, multicenter, RCT; sample size: 48; intervention: tacrolimus (0.05 mg/kg per d) versus supportive therapy; end point: remission proteinuria; follow-up: 18 mo Rate of remission at 18 mo higher in tacrolimus group (94%) versus control (35%); 50% relapse rate in tacrolimus group Tacrolimus (n=25) Supportive care (n=23)
Serious infection 0 0
Diarrhea 2 (8%) 0
Glucose intolerance 4 (16%) 2 (9%)
Thrombosis 0 0
Howman et al. (67) (membranous nephropathy) Prospective, multicenter, RCT (1998–2008); sample size: 108; intervention: prednisone/oral chlorambucil versus oral cyclosporine (5 mg/kg) versus supportive therapy; end point: 20% decline in kidney function; follow-up: 36 mo 20% kidney function decline was lower in prednisone/chlorambucil group (58%) Prednisone/chlorambucil (n=33) Cyclosporine (n=37) Supportive care (n=38)
SAEs 61% 49% 42%
Cytopenia 28 (85%) 5 (13%) 3 (8%)
Infection 3 (9%) 8 (22%) 0
Gastrointestinal 3 (9%) 3 (8%) 2 (5%)
Impaired glycemia 8 (24%) 1 (3%) 5 (13%)
Dahan et al. (41) (GEMRITUX, membranous nephropathy) Prospective, multicenter, RCT; sample size: 75; intervention: rituximab (375 mg/m2×2) versus control; end point: remission proteinuria; follow-up: 6 mo No difference in remission rates (rituximab 35% versus control 22%) Rituximab (n=37) Control (n=38)
SAEs 6 (16%) 5 (13%)
Serious infections 1 (3%) 0
Leukopenia 0 0
Cardiovascular events 4 (11%) 2 (5%)
Cancer 0 1 (3%)
Diarrhea 1 (3%) 0

RCT, randomized controlled trial; SAE, serious adverse event; GEMRITUX, Evaluate Rituximab Treatment for Idiopathic Membranous Nephropathy.

Osteoporosis

GCs are associated with an accelerated bone loss in the first 3–6 months, followed by a persistent decrease in bone formation for the duration of therapy. Fracture risk is related to both the peak and cumulative GC dose, and is increased even at chronic low doses of 5 mg prednisone per day. In patients on long-term steroids, the American College of Rheumatology 2017 guidelines recommend a fracture risk assessment using bone mineral density testing and an online risk assessment tool (FRAX; https://www.shef.ac.uk/FRAX/tool.jsp) for adults over 40 years of age (9). All patients should be treated with oral calcium (1000–1200 mg/d) and oral vitamin D (600–800 U/d). Oral bisphosphonates should be considered for those at moderate to high fracture risk (>10%–20% 10-year fracture risk) (9). Note in the treatment of glomerular disease, the duration of steroid therapy may be short, and in patients with CKD, bone density measurements may be less precise and the safety of bisphosphonate therapy uncertain.

Avascular Necrosis

Avascular necrosis (osteonecrosis), typically affecting the femoral head, is a severe complication of steroid therapy usually attributed to ischemia secondary to abnormalities of lipid metabolism, oxidative stress, and vascular injury. Younger patients with SLE are more prone to avascular necrosis, possibly because of the associated chronic inflammatory and procoagulant milieu. Hip pain typically occurs 2–3 years after the start of GC therapy, but may occur earlier. In contrast to GC-induced osteoporosis (see above), avascular necrosis rarely occurs in patients treated with peak doses of <20–30 mg prednisone per day.

Adrenal Suppression

Chronic GC use can suppress the hypothalamic-pituitary-adrenal axis by negative feedback on corticotropin releasing hormone and corticotropin. The hypothalamic-pituitary-adrenal axis may remain suppressed after GC therapy has been reduced or stopped, resulting in adrenal insufficiency. Chronic low cortisol levels may result in nausea and fatigue, but in conditions of acute stress such as surgery, an adrenal crisis may result. Stress-dose steroids may be considered perioperatively for patients at higher risk of adrenal insufficiency. The prevalence of biochemical adrenal insufficiency in observational studies varies widely (14%–63%) (10,11), but clinical insufficiency is uncommon. Some studies have found an association with steroid dose and duration (11), but surprisingly in others, no relationship was found (10). The risk of adrenal insufficiency is minimal if GC duration is <3 weeks. To avoid adrenal insufficiency, GCs are usually prescribed in the morning, alternate day dosing may be used, and with chronic use, although the dose can be tapered relatively rapidly to a physiologic GC level (approximately 7.5 mg/d), a slower taper is needed below this dose.

Mycophenolic Acid

Mycophenolate mofetil (MMF) and enteric-coated mycophenolate sodium are antiproliferative agents that inhibit inosine monophosphate dehydrogenase, the rate-limiting step of purine synthesis in both B and T lymphocytes. Both formulations are highly protein bound (97%) and free mycophenolic acid (MPA) levels may be increased with the hypoalbuminemia of nephrotic syndrome, potentially leading to more adverse events. Notably antacids/phosphate binders (up to 33%) and protein pump inhibitors (25% or more) may inhibit oral absorption and decrease both the efficacy and complication rate. MMF is widely used in glomerular disease, especially LN, but the greatest experience comes from its extensive use in solid organ transplantation. Nausea and diarrhea are the commonest side effects, which may be reduced in some patients with the enteric-coated formulation. MPA salts are strongly contraindicated in pregnancy because of the association with first trimester pregnancy loss and congenital malformations.

Infection Risk

Viral infections (herpes zoster, cytomegalovirus, herpes simplex) are a particular risk with MMF treatment and this risk is enhanced by bone marrow suppression and leucopenia. By contrast, HBV and HCV viral infections may not be greatly increased as MPA may suppress the expression of HBV surface antigen and HBV viral replication (12). MPA may also have a suppressive effect on P. jirovecii. In a review of four kidney transplant studies, PJP occurred in none of the 1068 patients exposed to MMF, compared with ten out of 563 with other immunosuppression (mostly azathioprine) (13).

MMF is widely used as both induction and maintenance therapy for LN (Table 3) (1416). In the North American Collaborative study, adverse events and severe infections were less common with MMF compared with CYC (16); however, this was not noted in the multicenter Aspreva Lupus Management Study (ALMS) (14). A Cochrane review of six studies (n=683) (17) and a more recent meta-analysis (18) reported no difference in major infections between MMF and CYC.

Table 3.

Selected randomized controlled trials of lupus nephritis

Study Study Design Efficacy Adverse Events
Houssiau et al. (72) (Eurolupus, lupus nephritis) Prospective, multicenter RCT; sample size: 90; intervention: high-dose IV CYC (eight doses 0.5–1 g/m2) versus low-dose IV CYC (6 doses 500 mg); end point: treatment failure; follow-up: 41 mo High-dose and low-dose IV CYC showed equivalent efficacy High-dose CYC (n=45) Low-dose CYC (n=45)
Severe infection 10 (22%) 5 (11%)
Other infection 7 (16%) 5 (11%)
Leucopenia 5 (11%) 5 (11%)
Amenorrhea 3 (7%) 3 (7%)
Houssiau et al. (32) (Eurolupus, 10-yr follow-up) Prospective, multicenter RCT; sample size: 90; intervention: high-dose IV CYC (eight doses 0.5–1 g/m2) versus low-dose IV CYC (six doses 500 mg); end point: treatment failure; follow-up: 10 yr Long-term results showed no difference in efficacy High-dose CYC (n=45) Low-dose CYC (n=45)
Cardiovascular events 4 (9%) 3 (7%)
Cancers 1 (2%) 6 (14%)
Successful pregnancy 9 (20%) 10 (22%)
Contreras et al. (73) (lupus nephritis) Prospective, single-center RCT (1996–2003); sample size: 59; intervention: IV CYC (0.5–1 g 3 monthly) versus oral AZA (1–3 mg/kg per d) versus oral MMF (1–3 g/d); end point: patient and kidney survival; follow-up: 72 mo Patient and kidney survival was lower in the IV CYC group Ratesa CYC (n=20) AZA (n=19) MMF (n=20)
Infections 77% 25% 32%
Major infections 25% 2% 2%
Leucopenia 10% 6% 2%
Amenorrhea 32% 8% 6%
Vomiting 55% 7% 14%
Diarrhea 12% 9% 12%
Death 4 (25%) 0 1 (5%)
Ginzler et al. (16) (lupus nephritis) Prospective, multicenter RCT; sample size: 140; intervention: oral MMF (3 g/d) versus IV CYC (0.5–1 g monthly), both arms high-dose glucocorticoids; end point: remission in proteinuria; follow-up: 6 mo MMF was superior in rate of remission compared with CYC (52.1% versus 30.4%) MMF (n=71) CYC (n=69)
Serious infections 1 (1%) 6 (9%)
Other infection 3 (4%) 5 (7%)
Upper GI symptoms 23 (32%) 25 (36%)
Diarrhea 15 (21%) 2 (3%)
Lymphopenia 18 (25%) 28 (40%)
Alopecia 0 8 (12%)
Amenorrhea 0 2 (3%)
Appel et al. (14) (ALMS, lupus nephritis) Prospective, multicenter RCT; sample size: 370; intervention: oral MMF (3 g/d) versus IV CYC (0.5–1 g monthly), both arms high-dose glucocorticoids; end point: decrease in proteinuria or serum creatinine; follow-up: 6 mo No difference in response rates (56.2% versus 53%) MMF (n=184) CYC (n=180)
SAE 51 (28%) 41 (23%)
Severe infection 22 (12%) 18 (10%)
Diarrhea 52 (28%) 23 (13%)
Withdrawal 24 (13%) 13 (7%)
Death 9 (5%) 5 (3%)
Dooley et al. (15) (ALMS maintenance, lupus nephritis) Prospective, multicenter RCT; sample size: 227; intervention: oral MMF (2 g) versus oral azathioprine (2 mg/kg per d); end point: treatment failure (death, ESKD, doubling creatinine, kidney flare, rescue therapy); follow-up: 36 mo MMF was superior to azathioprine (16.4% versus 32.4%) MMF (n=115) AZA (n=111)
SAE 27 (23%) 37 (33%)
Infection 91 (79%) 87 (78%)
Serious infection 11 (10%) 13 (12%)
Leucopenia 0 4 (4%)
Withdrawal 29 (25%) 44 (40%)
Death 0 1 (1%)
Houssiau et al. (74) (MAINTAIN, lupus nephritis) Prospective, multicenter RCT (2002–2006); sample size: 105; intervention: oral AZA (2 mg/kg) versus oral MMF (2 g/d); end point: time to relapse; follow-up: 3 yr No difference in time to relapse, or relapse rate AZA (n=52) MMF (n=53)
Any infection 14 (27%) 21 (40%)
Leucopenia 11 (21%) 2 (4%)
Nausea/diarrhea 8 (15%) 8 (15%)
Rovin et al. (76) (LUNAR, induction – lupus nephritis) Prospective, multicenter RCT (2006–2008); sample size: 144; intervention: RTX (500 mg ×2, repeat 6 mo) versus placebo, background prednisone and MMF (3 g/d); end point: kidney response; follow-up: 18 mo No difference in kidney response between the groups RTX/MMF (n=72) MMF (n=72)
SAE 29 (40%) 24 (33%)
Severe infection 14 (19%) 14 (19%)
Infusion reaction 12 (17%) 6 (8%)
Neutropenia 2 (3%) 1 (1%)
Death 2 (3%) 0
ACCESS trial group (76) (lupus nephritis) Prospective multicenter RCT; sample size: 134; intervention: IV abatacept (500–1000 mg monthly) versus placebo, on background Eurolupus regime of CYC+AZA; end point: complete kidney response; follow-up: 12 mo No difference between the groups (33% versus 31%) Abatacept+CYC/AZA (n=66) Placebo+CYC/AZA (n=68)
SAE 19 (29%) 20 (29%)
Severe infection 8 (12%) 5 (7%)
Any infection 31 (47%) 32 (47%)
Death 0 1 (1%)
Liu et al. (39) (lupus nephritis) Prospective multicenter RCT; sample size: 362; intervention: MMF (0.5 mg twice a day)+TAC (2 mg twice a day) versus IV CYC (0.75 mg/m2 monthly ×6); end point: complete remission; follow-up: 6 mo More patients in MMF/TAC group achieved complete remission (46% versus 26%) MMF+TAC (n=181) IV CYC (n=181)
SAE 27 (15%) 10 (6%)
Severe infection 24 (13%) 8 (4%)
Any infection 91 (50%) 95 (52%)
Leukopenia 1 (1%) 12 (7%)
Gastrointestinal 25 (14%) 43 (24%)
Menstrual abnormality 2 (1%) 7 (5%)
Hyperglycemia 5 (3%) 4 (3%)

RCT, randomized controlled trial; IV, intravenous; CYC, cyclophosphamide; AZA, azathioprine; MMF, mycophenolate mofetil; GI, gastrointestinal; ALMS, Aspreva Lupus Management Study; SAE, serious adverse event; MAINTAIN, Azathioprine versus mycophenolate mofetil for long-term immunosuppression in lupus nephritis; LUNAR, Lupus Nephritis Assessment with Rituximab; RTX, rituximab; ACCESS, Abatacept and Cyclophosphamide Combination Efficacy and Safety; TAC, tacrolimus.

a

% rates defined as 100×number of events/number of patient-years follow-up.

Small studies have suggested that therapeutic drug monitoring may improve the efficacy and decrease the incidence of side effects in glomerular disease (19), although the use of MPA levels as a guide to therapy has not been widely adopted.

Cyclophosphamide

CYC is an inactive prodrug that is metabolized to an active metabolite, phosphoramide mustard, and the bladder-toxic metabolite acrolein, by cytochrome P450 enzymes. Functional variants in cytochrome P450 enzyme genes have been associated with increased leukopenia (but also increased efficacy) in AAV (20). CYC is associated with multiple serious adverse effects, many of which occur early (bone marrow suppression, infection, hemorrhagic cystitis, infertility), but others may occur more than 10 years after therapy has stopped (malignancy).

Cytopenia and Infection

Early studies of LN and AAV used CYC for both induction and maintenance therapy, resulting in a high cumulative dose. Recent protocols seek to minimize CYC exposure by limiting therapy to induction, using lower doses and pulse intravenous therapy rather than daily oral administration and by switching early to rituximab (RTX) after one or two doses (21,22). Infection rates in recent studies of LN and AAV are described in Table 3. Lymphopenia (<1×109/L) resulting from CYC therapy correlates with an increase in opportunistic infections (23).

Cancer Risk

CYC is associated with an increase in malignances (mostly skin cancer, myeloid malignancies, and bladder cancer), because of direct DNA damage and reduced immune surveillance. Early studies, with typically high cumulative doses of CYC, showed a 2–2.4-fold increase in cancer rates, but more recent studies show a lower risk (24,25). In a Danish registry study with 10-year follow-up, patients with AAV treated with CYC (n=293) had a 27% incidence of malignancy with a standardized incidence ratio (SIR) of 1.9 compared with national cancer incidence rates, driven mostly by non-myeloma skin cancer (NMSC) (24). Notably, in the subgroup with a lower cumulative CYC exposure (<36 g), there was no increased cancer risk. In a review of four studies of AAV from the European Vasculitis Study Group (n=535), the use of CYC was associated with an increased SIR of 1.58, driven entirely by NMSC, although the median follow-up was short (5 years) (25). In a single-center series of patients with AAV (n=119) with a median 5.6 year follow-up, the SIR was 3.1, again by NMSC only, without an increase in bladder cancer or myeloid malignancy (26). Of note, this study did not find any increase in the cancer rate in patients treated with RTX, an alternative agent in the treatment of AAV. Bladder cancer is a specific concern after CYC use and may be related to the bladder toxic metabolite acrolein. A high frequency (4.8%–14%) was described in older studies using prolonged courses of CYC (50–100 g cumulative dose), but no increased risk was noted in two recent studies with lower cumulative drug exposure (<20–36 g) (24,27). The value of mesna for bladder protection is uncertain at the doses of CYC typically used for glomerular disease (28).

To put the cumulative dose thresholds into context, a patient weighing 70 kg, treated with a variety of common CYC protocols, would receive a cumulative dose much lower than 20 g: (modified Ponticelli regime for membranous nephropathy [13 g]; 6 month course for LN [6 g]; Eurolupus protocol for LN [3 g]; 3 month course of oral CYC, 2 mg/kg for minimal change disease, LN, or AAV [13 g]). Note in many of these studies, the role of the underlying disease, and the contribution from other immunosuppression is difficult to determine. For example, azathioprine is commonly used as maintenance therapy in AAV, and itself is associated with a three-fold increase in skin cancer risk.

Hemorrhagic Cystitis

This complication occurred commonly (12%–41%) with prolonged courses of high-dose oral CYC (50–100 g), and but is rare with intravenous CYC and with lower dose oral CYC (28). Mesna binds acrolein in the urine and has been used to prevent hemorrhagic cystitis, however, recent studies question its efficacy (28,29).

Fertility

CYC therapy is associated with decreased fertility in both men and women and is associated with cumulative drug exposure. In a review of the National Institutes of Health studies of LN, chronic amenorrhea occurred more commonly with higher dose (approximately 10–15 g) versus lower dose (approximately 5–6 g) CYC (36% versus 12%) (30). The rate also increased with age: <25 years (12%), 26–30 years (27%), and >31 years (62%). A second study of 84 women with AAV or LN treated with intravenous CYC (mean exposure 12 g) showed chronic amenorrhea in 70% of women over 35 years, but in 0% of those aged <26 years (31). The risk of amenorrhea seems to increase significantly after 10–15 g exposure, although no safe threshold has been demonstrated (30). In the Eurolupus study, much lower doses (3 g) were used and fertility rates were relatively preserved in this study (32). Gonadotropin-releasing hormone agonists (e.g., leuprolide) for ovarian suppression are often used for protection, but efficacy is debated (33), and oocyte or embryo cryopreservation should be considered if higher dose CYC is required.

In men, transient azoospermia is common, with recovery related to the cumulative dose. In patients treated for sarcoma, exposure to a cumulative dose <7.5 g/m2 (approximately 14 g for a 70 kg man) resulted in a 72% recovery, whereas only 11% recovered with doses >7.5 g/m2 (34). The risk of permanent azoospermia may be low for cumulative doses <12 g (equivalent to 2 mg/kg for 12 weeks) (35). Notably, gonadotropin-releasing hormone analogs have shown disappointing results for fertility protection in men and semen cryopreservation should be considered.

Calcineurin Inhibitors

The calcineurin inhibitors (CNIs) cyclosporine and tacrolimus are extensively used in the treatment of membranous nephropathy, minimal change disease, and FSGS, and more recently in the treatment of LN as part of multitarget therapy. CNIs inhibit T cell activation, but also have direct effects on the podocyte, stabilizing the actin cytoskeleton (36).

The side-effect profiles of these two agents are similar, although some adverse events are more common with cyclosporin (hirsutism, gingival hyperplasia, hyperuricemia, hypertension, hyperlipidemia) and others with tacrolimus (glucose intolerance, neurotoxicity). One of the major concerns of CNIs is nephrotoxicity, both as an early hemodynamic reversible acute injury and as a chronic progressive nephropathy. Tacrolimus is also associated with hyperkalemic metabolic acidosis and hypertension mediated by upregulation of the sodium chloride transporter in the distal convoluted tubule (37). Both CNIs have a narrow therapeutic window with marked variability in bioavailability and metabolism, and therapeutic drug monitoring is required. Genetic variation in genes encoding CYP3A4, CYP3A5, and P-glycoprotein contribute to the variable pharmacokinetics and have been associated with an risk of nephrotoxicity (38). Anemia and hypoalbuminemia may increase the unbound (active) fraction of CNI and increase side effects for a given whole-blood level.

In LN, recent trials have shown a benefit from multitarget therapy, adding a CNI to lower dose prednisone and mycophenolate. Liu et al. (39), showed that the MMF/tacrolimus group was superior to the intravenous CYC group in achieving a complete kidney response at 6 months (46% versus 26%); however, adverse events were increased, with more serious infections (Table 3). A novel CNI, voclosporin, has been studied in a phase IIb trial of LN (AURA-LV) (ClinicalTrials.gov: NCT02141672). The addition of voclosporin to prednisone/MMF increased remission rates at 48 weeks, but serious adverse events and mortality were increased in the voclosporin groups. The ongoing phase 3 study will provide further information on the adverse drug reactions with this combination.

CNIs continue to be widely used in the treatment of glomerular disease with drug interactions remaining one of the most challenging management issues. New extended release formulations of tacrolimus, and novel CNIs (e.g., voclosporin) may dramatically change the use of these agents.

Rituximab

This chimeric mAb to CD20 leads to depletion of circulating and tissue resident B cells, but not plasma cells (which lack the CD20 antigen). In the United States, it is approved by the US Food and Drug Administration for the treatment of AAV, but is widely used off-label in the treatment of other glomerular diseases. RTX has a prolonged duration of action. The drug can be found in the circulation for 3–6 months, and B cell depletion may be sustained for 6–12 months, and sometimes longer.

Infusion Reactions

Immediate hypersensitivity reactions are relatively common and were described in 28% of patients with idiopathic membranous nephropathy (40), but in a more recent study using appropriate prophylaxis, no severe infusion reactions were noted (41). These reactions are mediated by a cytokine release syndrome, not anaphylaxis, and manifest by severe flu-like symptoms (fever, chills, myalgias, shortness of breath). Pretreatment with acetaminophen, diphenhydramine, and methylprednisone, and starting with a slow infusion rate are commonly used to prevent this.

Infectious Complications

Infection rates after RTX therapy vary according to the indication for treatment, age, comorbidities, and the cumulative effect of other immunosuppressive agents. Hypogammaglobulinemia and late-onset neutropenia are additional risk factors. In the Rituximab in ANCA-associated vasculitis (RAVE) and Rituximab versus cyclophosphamide in ANCA-associated vasculitis (RITUXVAS) trials of AAV, severe infection was described in 7% and 18%, respectively, and somewhat surprisingly, did not differ from the CYC arm (22,42) (Table 4). By contrast, in a retrospective study (n=370) of autoimmune disorders (mostly rheumatoid arthritis, LN, and AAV), serious infection events occurred in only 3.7% (43). A recent study (n=98) of primary and secondary glomerular disease described an overall infection rate of 21.6 per 100 patient years (44). In other glomerular diseases, lower infection rates were described. No infections were described in cohorts of membranous nephropathy (n=100) (40), minimal change disease (n=17) (45), and only a single case in another cohort of membranous nephropathy (n=15) (46).

Table 4.

Selected randomized controlled trials of ANCA-associated vasculitis

Study Study Design Efficacy Adverse Events
Jayne et al. (68) (CYCAZAREM, maintenance AAV) Prospective, multicenter, RCT; sample size: 155; intervention: oral CYC (1.5 mg/kg) versus oral AZA (2 mg/kg); end point: relapse rate; follow-up: 18 mo No difference in relapse rate AZA (n=71) CYC (n=73)
SAEs 12 (28%) 15 (21%)
Any infection 13 (18%) 13 (14%)
Severe infection 4 (6%) 3 (4%)
Leukopenia 22 (31%) 35 (48%)
Cystitis 1 (1%) 3 (4%)
Amenorrhea 1 (1%) 2 (3%)
de Groot et al. (69) (CYCLOPS, AAV) Prospective, multicenter, RCT; sample size: 147; intervention: pulse IV CYC (15 mg/kg q2–3 weekly) versus oral CYC (2 mg/kg); end point: time to remission; follow-up: 18 mo No difference in time to remission Pulse IV CYC (n=76) Oral CYC (n=71)
SAE 19 (25%) 31 (44%)
Serious infections 7 (9%) 10 (14%)
Other infection 20 (26%) 21 (30%)
Leucopenia 20 (26%) 33 (46%)
Alopecia 0 2 (3%)
Amenorrhea 1 (1%) 0
New/worse diabetes 8 (11%) 8 11%)
Harper et al. (70) (CYCLOPS, AAV) Follow-up: median 43 mo Relapse rate increased in pulse IV CYC group (39.5% versus 20.8%) Pulse IV CYC (n=76) Oral CYC (n=71)
Severe infection 19 (25%) 15 (21%)
Cancer 8 (11%) 6 (8%)
Cardiovascular event 6 (8%) 3 (4%)
New onset diabetes 8 (11%) 5 (7%)
Fracture 6 (8%) 3 (4&)
Hiemstra et al. (71) (IMPROVE, maintenance AAV) Prospective, multicenter RCT (2002–2009); sample size: 156; intervention: oral AZA (2 mg/kg) versus oral MMF (2 g/d); end point: relapse-free survival; follow-up: 39 mo AZA was superior to MMF (relapse rate 38% versus 55%) AZA (n=80) MMF (n=76)
SAE 13 (16%) 8 (11%)
Any infection 17 (21%) 12 (16%)
Severe infection 8 (10%) 3 (4%)
Leucopenia 7 (9%) 4 (5%)
Cancer 3 (4%) 1 (1%)
Gastrointestinal 8 (10%) 8 (11%)
Stone et al. (22) (RAVE, induction AAV) Prospective, multicenter RCT (2004–2008); sample size: 197; intervention: RTX (375 mg/m2×4) versus oral CYC (2 mg/kg); end point: remission rate; follow-up: 6 mo No difference in primary end point; RTX superior in relapsing disease RTX (n=99) CYC (n=98)
SAE 11 (11%) 5 (5%)
Severe infection 7 (7%) 7 (7%)
Leucopenia 3 (3%) 10 10%)
Cancer 1 (1%) 1 (1%)
Gastrointestinal 8 (8%) 8 (8%)
Hemorrhagic cystitis 1 (1%) 1 (1%)
Venous thrombosis 6 (6%) 9 (9%)
Death 1 (1%) 2 (2%)
Jones et al. (42) (RITUXVAS, induction AAV) Prospective, multicenter RCT (2006–2007); sample size: 44; intervention: RTX (375 mg/m2×4) versus IV CYC (3–6 m), then maintenance AZA (2 mg/kg); end point: sustained remission rate; follow-up: 12 mo No difference in sustained remission rate RTX (n=33) CYC/AZA (n=11)
SAE 14 (42%) 4 (36%)
Any infection 12 (36%) 7 (64%)
Severe infection 6 (18%) 3 (27%)
Hypogammaglobulinemia 1 (3%) 0
Neutropenia 2 (6%) 1 (9%)
Infusion reaction 2 (6%) 0
Cancer 2 (6%) 0
Death 3 (9%) 1 (9%)
Guillevin et al. (21) (MAINRITSAN, maintenance AAV) Prospective, multicenter RCT (2008–2010); sample size: 115; intervention: RTX (500 mg×2, then 6 monthly) versus AZA 2 mg/kg, then 1.5 mg/kg); end point: major relapse; follow-up: 28 mo RTX was superior to AZA in preventing relapse (5% versus 29%) RTX (n=57) AZA (n=58)
SAE 25 (44%) 25 (43%)
Severe Infection 11 (19%) 8 (14%)
Pneumocystis jirovecii 1 (2%) 0
Leukopenia 0 6 (10%)
Cancer 0 2 (3%)
Death 0 2 (3%)

CYCAZAREM, Cyclophosphamide versus Azathioprine for Early Remission Phase of Vasculitis; AAV, ANCA-associated vasculitis; RCT, randomized controlled trial; CYC, cyclophosphamide; AZA, azathioprine; SAE, serious adverse event; CYCLOPS, Pulse versus daily oral cyclophosphamide for induction of remission in ANCA-associated vasculitis; IV, intravenous; IMPROVE, International Mycophenolate Mofetil Protocol to Reduce Outbreaks of Vasculitides; MMF, mycophenolate mofetil; RAVE, Rituximab in ANCA-Associated; RTX, rituximab; RITUXVAS, Rituximab versus cyclophosphamide in ANCA-associated vasculitis; MAINRITSAN, Rituximab versus azathioprine for maintenance in ANCA-associated vasculitis.

Certain opportunistic infections are of concern with RTX therapy. Progressive multifocal leukoencephalopathy (PML) is a rare, but often fatal, neurologic disease caused by reactivation of the John Cunningham polyomavirus, described in patients taking RTX, and a “black box” warning is included in the product label information in the United States. The large majority of cases are described in patients treated for lymphoma, who are confounded by other risk factors, including concomitant chemotherapy and the underlying hematologic malignancy (47). Multiple case reports of PML have also been described in patients with LN (47,48). In a database study of hospital discharges, PML occurred in 4 per 100,000 patients with SLE (49). The overall intensity of immunosuppression is critical and many cases of PML were described in patients with LN treated with alkylating agents, and not RTX.

PJP is more commonly associated with impaired T cell immunity (e.g., CYC, HIV infection), but is rarely described with RTX. Concomitant high corticosteroid use increases the risk. In AAV, one patient (n=57) developed PJP in the RTX arm of the MAINRITSAN study (Table 4) (21). In a cohort of patients with AAV (n=53), one fatal case of PJP was described 2 months after discontinuation of prophylaxis (50). Similarly, two cases of PJP were described in a cohort of 80 patients with AAV (51). In LN, the incidence in hospitalized patients may be similar, and is estimated at 1% (52).

Reactivation of HBV is listed as a product black box warning by the FDA, and screening for hepatitis B surface antigen and anti-hepatitis B core should be performed before initiation of treatment. If positive, antiviral prophylaxis with entecavir or tenofovir should be started and continued for a least 1 year after the last RTX dose, in view of late immune reconstitution.

Hypogammaglobulinemia

Hypogammaglobulinemia is typically a late complication of RTX therapy. In patients with AAV, severe hypogammaglobulinemia (<3–4 g/L) was described in 4%–10% of patients (5355). In these patients, infection risk is increased, especially pulmonary infections, and intravenous Ig replacement may be required. This complication appears more commonly in AAV, where risk factors include prolonged therapy with RTX, prior treatment with CYC, and low IgG levels before treatment. IgG levels should be checked in high-risk patients before treatment with RTX, and in those who develop infectious complications.

Late-Onset Neutropenia

This is defined as an absolute neutrophil count <1.5×109/L, occurring more than 1 month (usually 2–6 months) after the last RTX infusion. It occurs in patients with AAV, but rarely in other forms of primary glomerular disease. The incidence ranges from 6.5% to 11.9% in patients with AAV treated with RTX (5658). The exact mechanism is unclear, but bone marrow biopsy studies have documented myeloid maturation arrest (59). It is often asymptomatic and recovers spontaneously, but granulocyte colony-stimulating factor may be considered when associated with infection. Notably, after recovery, re-exposure to RTX rarely leads to recurrent neutropenia (56,57).

Eculizumab

Eculizumab is a humanized IgGκ mAb that blocks the cleavage of complement C5, inhibiting formation of the membrane attack complex (C5b-9) and release of the anaphylatoxin C5a. It is used in complement-mediated disorders such as C3 glomerulopathy and thrombotic microangiopathy. Notably, multiple new complement inhibitors are currently in development, with many acting more proximally in the complement cascade, and these will likely have very different adverse event profiles.

Infectious Complications

The clearance of encapsulated organisms is heavily dependent on C5b-9. Infection with Streptococcus pneumoniae or Haemophilus influenza is less common, possibly related to opsonization, but the risk of infection with Neisseria meningitidis is markedly increased, even in those who have been vaccinated (60). Starting at least 2 weeks before therapy, patients should receive two doses (2 months apart) of the quadrivalent meningococcal conjugate vaccine (MenACWY), with a booster dose every 5 years. Simultaneous immunization with a recombinant meningococcal B vaccine (MenB) is also recommended. Chemoprophylaxis with penicillin V or ciprofloxacin should be used for the first 4 weeks of therapy. Disseminated gonococcal infection has also been described and sexually active patients should be counseled. Upper respiratory tracts infections may also be more common (60).

Other Complications

Eculizumab is generally well tolerated and infusion reactions are rare. The development of low titer neutralizing antibodies to eculizumab is described (approximately 2%), but does not seem to interfere with clinical response.

Recommendations for Screening and Prophylaxis

Screening for Latent Infection

Before starting therapy with immunosuppression, screening for HBV, HCV, HIV, latent tuberculosis, and strongyloidiasis should be considered (Table 5). The patient should be assessed for evidence of mucosal candidiasis.

Table 5.

Screening and prophylaxis with immunosuppression in glomerular diseases

Assessment Recommendation
Infection screening
Hepatitis B HBsAg, anti-HBc antibody
Hepatitis C Anti-HCV antibody
HIV Anti-HIV antibody±HIV p24 antigen
Latent tuberculosis Chest x-ray, tuberculin skin test or IFN-γ release assay
Strongyloidiasis Anti-strongyloides antibody
Vaccination Per CDC guidelines (62) (avoid live vaccines during and within 4 wk of starting immunosuppression)
Infection prophylaxis
Pneumocystis jirovecii Cotrimoxazole or dapsone, atovaquone, inhaled pentamadine
Candida Clotrimazole troche or weekly fluconazole
Other prophylaxis
Osteoporosis Calcium and vitamin D; ±bisphosphonates
Gastroprophylaxis Histamine (H2) antagonist, proton pump inhibitor

HBsAg, hepatitis B surface antigen; HBc, hepatitis B core; HCV, hepatitis C virus; CDC, Centers for Disease Control.

Vaccination

The optimal time for vaccination is at least 4 weeks before starting treatment with immunosuppression, but this is not often possible in glomerular disease. Notably, after RTX or other anti-B cell therapies, the response to vaccination may be markedly attenuated. Vaccination should be performed according to latest Centers for Disease Control guidelines for immunocompromised patients (61). In general, patients should be vaccinated according to the standard Centers for Disease Control annual schedule, including inactivated pneumococcal and influenza vaccines, but avoiding live vaccines such as varicella. A new inactivated recombinant zoster vaccine became available in the United States in 2017.

Infection Prophylaxis

The question of when to use cotrimoxazole or other PJP chemoprophylaxis is difficult for clinicians caring for patients with glomerular disease. PJP has been described with GC therapy at doses as low as 16 mg/d for 8 weeks (62), but is more common at higher doses, and when GCs are used in combination with other immunosuppression. Impaired kidney function and hypoalbuminemia have been described as risk factors for PJP infection in patients with IgA nephropathy (63). Prophylaxis is typically used in AAV, and often in LN in the setting of high-dose steroids with additional immunosuppressive agents. In minimal change disease/FSGS/IgA nephropathy when high-dose GC monotherapy is used, prophylaxis should also be considered. Candida prophylaxis with clotrimazole troche or weekly oral fluconazole should also be considered in patients taking high-dose GCs (Table 5).

Other Prophylaxis

Recommendations for osteoporosis prophylaxis are described above. Patients treated with high-dose GCs are at risk for gastritis and peptic ulcer disease, and prophylaxis with histamine antagonists or proton pump inhibitors may be considered.

Disclosures

None.

Footnotes

Published online ahead of print. Publication date available at www.cjasn.org.

See related Commentary, “Commentary on Complications of Immunosuppressive Treatments for Glomerulonephritis,” on pages 1276–1277.

References

  • 1.Fardet L, Petersen I, Nazareth I: Common infections in patients prescribed systemic glucocorticoids in primary care: A population-based cohort study. PLoS Med 13: e1002024, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Youssef J, Novosad SA, Winthrop KL: Infection risk and safety of corticosteroid use. Rheum Dis Clin North Am 42: 157–176, ix–x, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lv J, Zhang H, Chen Y, Li G, Jiang L, Singh AK, Wang H: Combination therapy of prednisone and ACE inhibitor versus ACE-inhibitor therapy alone in patients with IgA nephropathy: A randomized controlled trial. Am J Kidney Dis 53: 26–32, 2009 [DOI] [PubMed] [Google Scholar]
  • 4.Manno C, Torres DD, Rossini M, Pesce F, Schena FP: Randomized controlled clinical trial of corticosteroids plus ACE-inhibitors with long-term follow-up in proteinuric IgA nephropathy. Nephrol Dial Transplant 24: 3694–3701, 2009 [DOI] [PubMed] [Google Scholar]
  • 5.Pozzi C, Bolasco PG, Fogazzi GB, Andrulli S, Altieri P, Ponticelli C, Locatelli F: Corticosteroids in IgA nephropathy: A randomised controlled trial. Lancet 353: 883–887, 1999 [DOI] [PubMed] [Google Scholar]
  • 6.Rauen T, Fitzner C, Eitner F, Sommerer C, Zeier M, Otte B, Panzer U, Peters H, Benck U, Mertens PR, Kuhlmann U, Witzke O, Gross O, Vielhauer V, Mann JFE, Hilgers RD, Floege J: Effects of two immunosuppressive treatment protocols for IgA nephropathy. J Am Soc Nephrol 29: 317–325, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lv J, Zhang H, Wong MG, Jardine MJ, Hladunewich M, Jha V, Monaghan H, Zhao M, Barbour S, Reich H, Cattran D, Glassock R, Levin A, Wheeler D, Woodward M, Billot L, Chan TM, Liu ZH, Johnson DW, Cass A, Feehally J, Floege J, Remuzzi G, Wu Y, Agarwal R, Wang HY, Perkovic V; TESTING Study Group: Effect of oral methylprednisolone on clinical outcomes in patients with IgA nephropathy: The TESTING randomized clinical trial. JAMA 318: 432–442, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Flanc RS, Roberts MA, Strippoli GF, Chadban SJ, Kerr PG, Atkins RC: Treatment of diffuse proliferative lupus nephritis: A meta-analysis of randomized controlled trials. Am J Kidney Dis 43: 197–208, 2004 [DOI] [PubMed] [Google Scholar]
  • 9.Buckley L, Guyatt G, Fink HA, Cannon M, Grossman J, Hansen KE, Humphrey MB, Lane NE, Magrey M, Miller M, Morrison L, Rao M, Byun Robinson A, Saha S, Wolver S, Bannuru RR, Vaysbrot E, Osani M, Turgunbaev M, Miller AS, McAlindon T: 2017 American college of rheumatology guideline for the prevention and treatment of glucocorticoid-induced osteoporosis. Arthritis Care Res (Hoboken) 69: 1095–1110, 2017 [DOI] [PubMed] [Google Scholar]
  • 10.Jamilloux Y, Liozon E, Pugnet G, Nadalon S, Heang Ly K, Dumonteil S, Gondran G, Fauchais AL, Vidal E: Recovery of adrenal function after long-term glucocorticoid therapy for giant cell arteritis: A cohort study. PLoS One 8: e68713, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Schlaghecke R, Kornely E, Santen RT, Ridderskamp P: The effect of long-term glucocorticoid therapy on pituitary-adrenal responses to exogenous corticotropin-releasing hormone. N Engl J Med 326: 226–230, 1992 [DOI] [PubMed] [Google Scholar]
  • 12.Wu J, Xie HY, Jiang GP, Xu X, Zheng SS: The effect of mycophenolate acid on hepatitis B virus replication in vitro. Hepatobiliary Pancreat Dis Int 2: 410–413, 2003 [PubMed] [Google Scholar]
  • 13.Husain S, Singh N: The impact of novel immunosuppressive agents on infections in organ transplant recipients and the interactions of these agents with antimicrobials. Clin Infect Dis 35: 53–61, 2002 [DOI] [PubMed] [Google Scholar]
  • 14.Appel GB, Contreras G, Dooley MA, Ginzler EM, Isenberg D, Jayne D, Li LS, Mysler E, Sánchez-Guerrero J, Solomons N, Wofsy D; Aspreva Lupus Management Study Group: Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J Am Soc Nephrol 20: 1103–1112, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dooley MA, Jayne D, Ginzler EM, Isenberg D, Olsen NJ, Wofsy D, Eitner F, Appel GB, Contreras G, Lisk L, Solomons N; ALMS Group: Mycophenolate versus azathioprine as maintenance therapy for lupus nephritis. N Engl J Med 365: 1886–1895, 2011 [DOI] [PubMed] [Google Scholar]
  • 16.Ginzler EM, Dooley MA, Aranow C, Kim MY, Buyon J, Merrill JT, Petri M, Gilkeson GS, Wallace DJ, Weisman MH, Appel GB: Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis. N Engl J Med 353: 2219–2228, 2005 [DOI] [PubMed] [Google Scholar]
  • 17.Henderson LK, Masson P, Craig JC, Roberts MA, Flanc RS, Strippoli GF, Webster AC: Induction and maintenance treatment of proliferative lupus nephritis: A meta-analysis of randomized controlled trials. Am J Kidney Dis 61: 74–87, 2013 [DOI] [PubMed] [Google Scholar]
  • 18.Singh JA, Hossain A, Kotb A, Wells G: Risk of serious infections with immunosuppressive drugs and glucocorticoids for lupus nephritis: A systematic review and network meta-analysis. BMC Med 14: 137, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zabotti A, Baraldo M, Quartuccio L, Sacco S, De Marchi G, De Vita S: Optimizing the dose of mycophenolate mofetil for the maintenance treatment of lupus nephritis by therapeutic drug monitoring. Clin Rheumatol 34: 171–174, 2015 [DOI] [PubMed] [Google Scholar]
  • 20.Schirmer JH, Bremer JP, Moosig F, Holle JU, Lamprecht P, Wieczorek S, Haenisch S, Cascorbi I: Cyclophosphamide treatment-induced leukopenia rates in ANCA-associated vasculitis are influenced by variant CYP450 2C9 genotypes. Pharmacogenomics 17: 367–374, 2016 [DOI] [PubMed] [Google Scholar]
  • 21.Guillevin L, Pagnoux C, Karras A, Khouatra C, Aumaître O, Cohen P, Maurier F, Decaux O, Ninet J, Gobert P, Quémeneur T, Blanchard-Delaunay C, Godmer P, Puéchal X, Carron PL, Hatron PY, Limal N, Hamidou M, Ducret M, Daugas E, Papo T, Bonnotte B, Mahr A, Ravaud P, Mouthon L; French Vasculitis Study Group: Rituximab versus azathioprine for maintenance in ANCA-associated vasculitis. N Engl J Med 371: 1771–1780, 2014 [DOI] [PubMed] [Google Scholar]
  • 22.Stone JH, Merkel PA, Spiera R, Seo P, Langford CA, Hoffman GS, Kallenberg CG, St Clair EW, Turkiewicz A, Tchao NK, Webber L, Ding L, Sejismundo LP, Mieras K, Weitzenkamp D, Ikle D, Seyfert-Margolis V, Mueller M, Brunetta P, Allen NB, Fervenza FC, Geetha D, Keogh KA, Kissin EY, Monach PA, Peikert T, Stegeman C, Ytterberg SR, Specks U; RAVE-ITN Research Group: Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med 363: 221–232, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Goupil R, Brachemi S, Nadeau-Fredette AC, Déziel C, Troyanov Y, Lavergne V, Troyanov S: Lymphopenia and treatment-related infectious complications in ANCA-associated vasculitis. Clin J Am Soc Nephrol 8: 416–423, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Faurschou M, Mellemkjaer L, Voss A, Keller KK, Hansen IT, Baslund B: Prolonged risk of specific malignancies following cyclophosphamide therapy among patients with granulomatosis with polyangiitis. Rheumatology (Oxford) 54: 1345–1350, 2015 [DOI] [PubMed] [Google Scholar]
  • 25.Heijl C, Harper L, Flossmann O, Stücker I, Scott DG, Watts RA, Höglund P, Westman K, Mahr A; European Vasculitis Study Group (EUVAS): Incidence of malignancy in patients treated for antineutrophil cytoplasm antibody-associated vasculitis: Follow-up data from European Vasculitis Study Group clinical trials. Ann Rheum Dis 70: 1415–1421, 2011 [DOI] [PubMed] [Google Scholar]
  • 26.van Daalen EE, Rizzo R, Kronbichler A, Wolterbeek R, Bruijn JA, Jayne DR, Bajema IM, Rahmattulla C: Effect of rituximab on malignancy risk in patients with ANCA-associated vasculitis. Ann Rheum Dis 76: 1064–1069, 2017 [DOI] [PubMed] [Google Scholar]
  • 27.Travis LB, Curtis RE, Glimelius B, Holowaty EJ, Van Leeuwen FE, Lynch CF, Hagenbeek A, Stovall M, Banks PM, Adami J, Gospodarowicz MK, Wacholder S, Inskip PD, Tucker MA, Boice JD: Bladder and kidney cancer following cyclophosphamide therapy for non-Hodgkin’s lymphoma. J Natl Cancer Inst 87: 524–530, 1995 [DOI] [PubMed] [Google Scholar]
  • 28.Monach PA, Arnold LM, Merkel PA: Incidence and prevention of bladder toxicity from cyclophosphamide in the treatment of rheumatic diseases: A data-driven review. Arthritis Rheum 62: 9–21, 2010 [DOI] [PubMed] [Google Scholar]
  • 29.Yilmaz N, Emmungil H, Gucenmez S, Ozen G, Yildiz F, Balkarli A, Kimyon G, Coskun BN, Dogan I, Pamuk ON, Yasar S, Cetin GY, Yazici A, Ergulu Esmen S, Cagatay Y, Yilmaz S, Cefle A, Sayarlioglu M, Kasifoglu T, Karadag O, Pehlivan Y, Dalkilic E, Kisacik B, Cobankara V, Erken E, Direskeneli H, Aksu K, Yavuz S: Incidence of cyclophosphamide-induced urotoxicity and protective effect of mesna in rheumatic diseases. J Rheumatol 42: 1661–1666, 2015 [DOI] [PubMed] [Google Scholar]
  • 30.Boumpas DT, Austin HA 3rd, Vaughan EM, Yarboro CH, Klippel JH, Balow JE: Risk for sustained amenorrhea in patients with systemic lupus erythematosus receiving intermittent pulse cyclophosphamide therapy. Ann Intern Med 119: 366–369, 1993 [DOI] [PubMed] [Google Scholar]
  • 31.Huong DL, Amoura Z, Duhaut P, Sbai A, Costedoat N, Wechsler B, Piette JC: Risk of ovarian failure and fertility after intravenous cyclophosphamide. A study in 84 patients. J Rheumatol 29: 2571–2576, 2002 [PubMed] [Google Scholar]
  • 32.Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, de Ramon Garrido E, Danieli MG, Abramovicz D, Blockmans D, Cauli A, Direskeneli H, Galeazzi M, Gül A, Levy Y, Petera P, Popovic R, Petrovic R, Sinico RA, Cattaneo R, Font J, Depresseux G, Cosyns JP, Cervera R: The 10-year follow-up data of the Euro-Lupus Nephritis Trial comparing low-dose and high-dose intravenous cyclophosphamide. Ann Rheum Dis 69: 61–64, 2010 [DOI] [PubMed] [Google Scholar]
  • 33.Demeestere I, Brice P, Peccatori FA, Kentos A, Dupuis J, Zachee P, Casasnovas O, Van Den Neste E, Dechene J, De Maertelaer V, Bron D, Englert Y: No evidence for the benefit of gonadotropin-releasing hormone agonist in preserving ovarian function and fertility in lymphoma survivors treated with chemotherapy: Final long-term report of a prospective randomized trial. J Clin Oncol 34: 2568–2574, 2016 [DOI] [PubMed] [Google Scholar]
  • 34.Meistrich ML, Wilson G, Brown BW, da Cunha MF, Lipshultz LI: Impact of cyclophosphamide on long-term reduction in sperm count in men treated with combination chemotherapy for Ewing and soft tissue sarcomas. Cancer 70: 2703–2712, 1992 [DOI] [PubMed] [Google Scholar]
  • 35.Wetzels JF: Cyclophosphamide-induced gonadal toxicity: A treatment dilemma in patients with lupus nephritis? Neth J Med 62: 347–352, 2004 [PubMed] [Google Scholar]
  • 36.Faul C, Donnelly M, Merscher-Gomez S, Chang YH, Franz S, Delfgaauw J, Chang JM, Choi HY, Campbell KN, Kim K, Reiser J, Mundel P: The actin cytoskeleton of kidney podocytes is a direct target of the antiproteinuric effect of cyclosporine A. Nat Med 14: 931–938, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hoorn EJ, Walsh SB, McCormick JA, Fürstenberg A, Yang CL, Roeschel T, Paliege A, Howie AJ, Conley J, Bachmann S, Unwin RJ, Ellison DH: The calcineurin inhibitor tacrolimus activates the renal sodium chloride cotransporter to cause hypertension. Nat Med 17: 1304–1309, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hesselink DA, Bouamar R, Elens L, van Schaik RH, van Gelder T: The role of pharmacogenetics in the disposition of and response to tacrolimus in solid organ transplantation. Clin Pharmacokinet 53: 123–139, 2014 [DOI] [PubMed] [Google Scholar]
  • 39.Liu Z, Zhang H, Liu Z, Xing C, Fu P, Ni Z, Chen J, Lin H, Liu F, He Y, He Y, Miao L, Chen N, Li Y, Gu Y, Shi W, Hu W, Liu Z, Bao H, Zeng C, Zhou M: Multitarget therapy for induction treatment of lupus nephritis: A randomized trial. Ann Intern Med 162: 18–26, 2015 [DOI] [PubMed] [Google Scholar]
  • 40.Ruggenenti P, Cravedi P, Chianca A, Perna A, Ruggiero B, Gaspari F, Rambaldi A, Marasà M, Remuzzi G: Rituximab in idiopathic membranous nephropathy. J Am Soc Nephrol 23: 1416–1425, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Dahan K, Debiec H, Plaisier E, Cachanado M, Rousseau A, Wakselman L, Michel PA, Mihout F, Dussol B, Matignon M, Mousson C, Simon T, Ronco P; GEMRITUX Study Group: Rituximab for severe membranous nephropathy: A 6-month trial with extended follow-up. J Am Soc Nephrol 28: 348–358, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jones RB, Tervaert JW, Hauser T, Luqmani R, Morgan MD, Peh CA, Savage CO, Segelmark M, Tesar V, van Paassen P, Walsh D, Walsh M, Westman K, Jayne DR; European Vasculitis Study Group: Rituximab versus cyclophosphamide in ANCA-associated renal vasculitis. N Engl J Med 363: 211–220, 2010 [DOI] [PubMed] [Google Scholar]
  • 43.Tony HP, Burmester G, Schulze-Koops H, Grunke M, Henes J, Kötter I, Haas J, Unger L, Lovric S, Haubitz M, Fischer-Betz R, Chehab G, Rubbert-Roth A, Specker C, Weinerth J, Holle J, Müller-Ladner U, König R, Fiehn C, Burgwinkel P, Budde K, Sörensen H, Meurer M, Aringer M, Kieseier B, Erfurt-Berge C, Sticherling M, Veelken R, Ziemann U, Strutz F, von Wussow P, Meier FM, Hunzelmann N, Schmidt E, Bergner R, Schwarting A, Eming R, Hertl M, Stadler R, Schwarz-Eywill M, Wassenberg S, Fleck M, Metzler C, Zettl U, Westphal J, Heitmann S, Herzog AL, Wiendl H, Jakob W, Schmidt E, Freivogel K, Dörner T; GRAID investigators: Safety and clinical outcomes of rituximab therapy in patients with different autoimmune diseases: Experience from a national registry (GRAID). Arthritis Res Ther 13: R75, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Trivin C, Tran A, Moulin B, Choukroun G, Gatault P, Courivaud C, Augusto JF, Ficheux M, Vigneau C, Thervet E, Karras A: Infectious complications of a rituximab-based immunosuppressive regimen in patients with glomerular disease. Clin Kidney J 10: 461–469, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Munyentwali H, Bouachi K, Audard V, Remy P, Lang P, Mojaat R, Deschênes G, Ronco PM, Plaisier EM, Dahan KY: Rituximab is an efficient and safe treatment in adults with steroid-dependent minimal change disease. Kidney Int 83: 511–516, 2013 [DOI] [PubMed] [Google Scholar]
  • 46.Fervenza FC, Abraham RS, Erickson SB, Irazabal MV, Eirin A, Specks U, Nachman PH, Bergstralh EJ, Leung N, Cosio FG, Hogan MC, Dillon JJ, Hickson LJ, Li X, Cattran DC; Mayo Nephrology Collaborative Group: Rituximab therapy in idiopathic membranous nephropathy: A 2-year study. Clin J Am Soc Nephrol 5: 2188–2198, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Carson KR, Evens AM, Richey EA, Habermann TM, Focosi D, Seymour JF, Laubach J, Bawn SD, Gordon LI, Winter JN, Furman RR, Vose JM, Zelenetz AD, Mamtani R, Raisch DW, Dorshimer GW, Rosen ST, Muro K, Gottardi-Littell NR, Talley RL, Sartor O, Green D, Major EO, Bennett CL: Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: A report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 113: 4834–4840, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Molloy ES, Calabrese CM, Calabrese LH: The risk of progressive multifocal leukoencephalopathy in the biologic era: Prevention and management. Rheum Dis Clin North Am 43: 95–109, 2017 [DOI] [PubMed] [Google Scholar]
  • 49.Molloy ES, Calabrese LH: Progressive multifocal leukoencephalopathy: A national estimate of frequency in systemic lupus erythematosus and other rheumatic diseases. Arthritis Rheum 60: 3761–3765, 2009 [DOI] [PubMed] [Google Scholar]
  • 50.Cartin-Ceba R, Golbin JM, Keogh KA, Peikert T, Sánchez-Menéndez M, Ytterberg SR, Fervenza FC, Specks U: Rituximab for remission induction and maintenance in refractory granulomatosis with polyangiitis (Wegener’s): Ten-year experience at a single center. Arthritis Rheum 64: 3770–3778, 2012 [DOI] [PubMed] [Google Scholar]
  • 51.Charles P, Néel A, Tieulié N, Hot A, Pugnet G, Decaux O, Marie I, Khellaf M, Kahn JE, Karras A, Ziza JM, Deligny C, Tchérakian C, Guillevin L; French Vasculitis Study Group: Rituximab for induction and maintenance treatment of ANCA-associated vasculitides: A multicentre retrospective study on 80 patients. Rheumatology (Oxford) 53: 532–539, 2014 [DOI] [PubMed] [Google Scholar]
  • 52.Weng CT, Liu MF, Weng MY, Lee NY, Wang MC, Lin WC, Ou CY, Lai WW, Hsu SC, Chao SC, Chung TJ, Lee CT, Shieh CC, Wang JY, Wang CR: Pneumocystis jirovecii pneumonia in systemic lupus erythematosus from southern Taiwan. J Clin Rheumatol 19: 252–258, 2013 [DOI] [PubMed] [Google Scholar]
  • 53.Alberici F, Smith RM, Jones RB, Roberts DM, Willcocks LC, Chaudhry A, Smith KG, Jayne DR: Long-term follow-up of patients who received repeat-dose rituximab as maintenance therapy for ANCA-associated vasculitis. Rheumatology (Oxford) 54: 1153–1160, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Marco H, Smith RM, Jones RB, Guerry MJ, Catapano F, Burns S, Chaudhry AN, Smith KG, Jayne DR: The effect of rituximab therapy on immunoglobulin levels in patients with multisystem autoimmune disease. BMC Musculoskelet Disord 15: 178, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Venhoff N, Effelsberg NM, Salzer U, Warnatz K, Peter HH, Lebrecht D, Schlesier M, Voll RE, Thiel J: Impact of rituximab on immunoglobulin concentrations and B cell numbers after cyclophosphamide treatment in patients with ANCA-associated vasculitides. PLoS One 7: e37626, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Knight A, Sundström Y, Börjesson O, Bruchfeld A, Malmström V, Gunnarsson I: Late-onset neutropenia after rituximab in ANCA-associated vasculitis. Scand J Rheumatol 45: 404–407, 2016 [DOI] [PubMed] [Google Scholar]
  • 57.Monaco WE, Jones JD, Rigby WF: Rituximab associated late-onset neutropenia-a rheumatology case series and review of the literature. Clin Rheumatol 35: 2457–2462, 2016 [DOI] [PubMed] [Google Scholar]
  • 58.Pendergraft WF 3rd, Cortazar FB, Wenger J, Murphy AP, Rhee EP, Laliberte KA, Niles JL: Long-term maintenance therapy using rituximab-induced continuous B-cell depletion in patients with ANCA vasculitis. Clin J Am Soc Nephrol 9: 736–744, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Tesfa D, Gelius T, Sander B, Kimby E, Fadeel B, Palmblad J, Hägglund H: Late-onset neutropenia associated with rituximab therapy: Evidence for a maturation arrest at the (pro)myelocyte stage of granulopoiesis. Med Oncol 25: 374–379, 2008 [DOI] [PubMed] [Google Scholar]
  • 60.Benamu E, Montoya JG: Infections associated with the use of eculizumab: Recommendations for prevention and prophylaxis. Curr Opin Infect Dis 29: 319–329, 2016 [DOI] [PubMed] [Google Scholar]
  • 61.Rubin LG, Levin MJ, Ljungman P, Davies EG, Avery R, Tomblyn M, Bousvaros A, Dhanireddy S, Sung L, Keyserling H, Kang I; Infectious Diseases Society of America: 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin Infect Dis 58: e44–e100, 2014 [DOI] [PubMed] [Google Scholar]
  • 62.Yale SH, Limper AH: Pneumocystis carinii pneumonia in patients without acquired immunodeficiency syndrome: Associated illness and prior corticosteroid therapy. Mayo Clin Proc 71: 5–13, 1996 [DOI] [PubMed] [Google Scholar]
  • 63.Ye WL, Tang N, Wen YB, Li H, Li MX, Du B, Li XM: Underlying renal insufficiency: The pivotal risk factor for Pneumocystis jirovecii pneumonia in immunosuppressed patients with non-transplant glomerular disease. Int Urol Nephrol 48: 1863–1871, 2016 [DOI] [PubMed] [Google Scholar]
  • 64.Rauen T, Eitner F, Fitzner C, Sommerer C, Zeier M, Otte B, Panzer U, Peters H, Benck U, Mertens PR, Kuhlmann U, Witzke O, Gross O, Vielhauer V, Mann JF, Hilgers RD, Floege J; STOP-IgAN Investigators: Intensive supportive care plus immunosuppression in IgA nephropathy. N Engl J Med 373: 2225–2236, 2015 [DOI] [PubMed] [Google Scholar]
  • 65.Hou JH, Le WB, Chen N, Wang WM, Liu ZS, Liu D, Chen JH, Tian J, Fu P, Hu ZX, Zeng CH, Liang SS, Zhou ML, Zhang HT, Liu ZH: Mycophenolate mofetil combined with prednisone versus full-dose prednisone in IgA nephropathy with active proliferative lesions: A randomized controlled trial. Am J Kidney Dis 69: 788–795, 2017 [DOI] [PubMed] [Google Scholar]
  • 66.Praga M, Barrio V, Juárez GF, Luño J; Grupo Español de Estudio de la Nefropatía Membranosa: Tacrolimus monotherapy in membranous nephropathy: A randomized controlled trial. Kidney Int 71: 924–930, 2007 [DOI] [PubMed] [Google Scholar]
  • 67.Howman A, Chapman TL, Langdon MM, Ferguson C, Adu D, Feehally J, Gaskin GJ, Jayne DR, O’Donoghue D, Boulton-Jones M, Mathieson PW: Immunosuppression for progressive membranous nephropathy: A UK randomised controlled trial. Lancet 381: 744–751, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Jayne D, Rasmussen N, Andrassy K, Bacon P, Tervaert JW, Dadoniené J, Ekstrand A, Gaskin G, Gregorini G, de Groot K, Gross W, Hagen EC, Mirapeix E, Pettersson E, Siegert C, Sinico A, Tesar V, Westman K, Pusey C; European Vasculitis Study Group: A randomized trial of maintenance therapy for vasculitis associated with antineutrophil cytoplasmic autoantibodies. N Engl J Med 349: 36–44, 2003 [DOI] [PubMed] [Google Scholar]
  • 69.de Groot K, Harper L, Jayne DR, Flores Suarez LF, Gregorini G, Gross WL, Luqmani R, Pusey CD, Rasmussen N, Sinico RA, Tesar V, Vanhille P, Westman K, Savage CO; EUVAS (European Vasculitis Study Group): Pulse versus daily oral cyclophosphamide for induction of remission in antineutrophil cytoplasmic antibody-associated vasculitis: A randomized trial. Ann Intern Med 150: 670–680, 2009 [DOI] [PubMed] [Google Scholar]
  • 70.Harper L, Morgan MD, Walsh M, Hoglund P, Westman K, Flossmann O, Tesar V, Vanhille P, de Groot K, Luqmani R, Flores-Suarez LF, Watts R, Pusey C, Bruchfeld A, Rasmussen N, Blockmans D, Savage CO, Jayne D; EUVAS investigators: Pulse versus daily oral cyclophosphamide for induction of remission in ANCA-associated vasculitis: Long-term follow-up. Ann Rheum Dis 71: 955–960, 2012 [DOI] [PubMed] [Google Scholar]
  • 71.Hiemstra TF, Walsh M, Mahr A, Savage CO, de Groot K, Harper L, Hauser T, Neumann I, Tesar V, Wissing KM, Pagnoux C, Schmitt W, Jayne DR; European Vasculitis Study Group (EUVAS): Mycophenolate mofetil vs azathioprine for remission maintenance in antineutrophil cytoplasmic antibody-associated vasculitis: A randomized controlled trial. JAMA 304: 2381–2388, 2010 [DOI] [PubMed] [Google Scholar]
  • 72.Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, Garrido Ed ER, Danieli MG, Abramovicz D, Blockmans D, Mathieu A, Direskeneli H, Galeazzi M, Gül A, Levy Y, Petera P, Popovic R, Petrovic R, Sinico RA, Cattaneo R, Font J, Depresseux G, Cosyns JP, Cervera R: Immunosuppressive therapy in lupus nephritis: The Euro-Lupus Nephritis Trial, a randomized trial of low-dose versus high-dose intravenous cyclophosphamide. Arthritis Rheum 46: 2121–2131, 2002 [DOI] [PubMed] [Google Scholar]
  • 73.Contreras G, Pardo V, Leclercq B, Lenz O, Tozman E, O’Nan P, Roth D: Sequential therapies for proliferative lupus nephritis. N Engl J Med 350: 971–980, 2004 [DOI] [PubMed] [Google Scholar]
  • 74.Houssiau FA, D’Cruz D, Sangle S, Remy P, Vasconcelos C, Petrovic R, Fiehn C, de Ramon Garrido E, Gilboe IM, Tektonidou M, Blockmans D, Ravelingien I, le Guern V, Depresseux G, Guillevin L, Cervera R; MAINTAIN Nephritis Trial Group: Azathioprine versus mycophenolate mofetil for long-term immunosuppression in lupus nephritis: Results from the MAINTAIN Nephritis Trial. Ann Rheum Dis 69: 2083–2089, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, Maciuca R, Zhang D, Garg JP, Brunetta P, Appel G; LUNAR Investigator Group: Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: The Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum 64: 1215–1226, 2012 [DOI] [PubMed] [Google Scholar]
  • 76.ACCESS Trial Group: Treatment of lupus nephritis with abatacept: The Abatacept and Cyclophosphamide Combination Efficacy and Safety Study. Arthritis Rheumatol 66: 3096–3104, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Journal of the American Society of Nephrology : CJASN are provided here courtesy of American Society of Nephrology

RESOURCES