Recently, the term monoclonal gammopathy of renal significance (MGRS) was introduced to distinguish monoclonal gammopathies that result in the development of kidney disease from those that are benign. By definition, patients with MGRS have B-cell clones that do not meet the definition of multiple myeloma or lymphoma. Nevertheless, these clones produce monoclonal proteins that are capable of injuring the kidney resulting in permanent damage. Except for immunoglobulin light chain amyloidosis with heart involvement in which death can be rapid, treatment of MGRS is often indicated more to preserve kidney function and prevent recurrence after kidney transplantation rather than the prolongation of life. Clinical trials are rare for MGRS-related kidney diseases, except in immunoglobulin light chain amyloidosis. Treatment recommendations are therefore based on the clinical data obtained from treatment of the clonal disorder in its malignant state. The establishment of these treatment recommendations is important until data can be obtained by clinical trials of MGRS-related kidney diseases.

Monoclonal gammopathy of renal significance (MGRS) is defined by the causal relationship between a small B-cell clone and renal disease, usually through deposition of the secreted monoclonal immunoglobulin (MIg) or a fragment thereof.1  The spectrum of MGRS is evolving, with the recent description of novel entities. With the exception of immunoglobulin light chain (AL) amyloidosis, few studies have focused on therapeutic issues. The International Kidney and Monoclonal Gammopathy Research Group initiated a collaborative effort aimed at delineating treatment strategies.

The B-cell clone corresponds to the definition of a dangerous small B-cell clone, suggesting that its deleterious consequences are not directly related to cellular proliferation but to other mechanisms, such as MIg deposition.2  Accordingly, myeloma cast nephropathy, which almost invariably complicates high tumor mass myeloma, should not be included in MGRS. Most MGRS are due to deposition of the MIg fragment with distinct localization and pattern of ultrastructural organization. This results in glomerulopathies with organized deposits, either fibrillar (AL, immunoglobulin heavy chain [AH] and immunoglobulin light and heavy chain [ALH] amyloidosis), microtubular (type I and type II cryoglobulinemias, immunotactoid glomerulopathy [ITG]), or nonorganized deposits (Randall type monoclonal immunoglobulin deposition disease [MIDD] and non-Randall type proliferative glomerulonephritis with monoclonal immunoglobulin deposits [PGNMID]). MGRS also includes tubular disorders such as Fanconi syndrome (FS).3,4  Nonamyloid fibrillary glomerulonephritis should not be considered as MGRS because it is nearly always characterized by polyclonal IgG deposits without a detectable clonal B-cell disorder.5,6  In MGRS, deposits of different ultrastructural patterns derived from the same MIg can occur. For instance, fibrillar AL and amorphous Randall-type deposits can coexist in the same patient.7  In most cases, the overall survival of patients with MGRS is significantly better than that of multiple myeloma, but the renal outcomes are not.1  The exception is patients with AL amyloidosis, particularly those with cardiac involvement in which death can occur rapidly.8  Many patients who develop end-stage renal disease (ESRD) are often not considered for kidney transplantation due to their high rate of recurrence.

In a patient in whom MGRS is suspected, it is essential to assess the characteristics of monoclonal gammopathy, particularly its isotype and whether it corresponds to an overt lymphoid and/or plasmacytic disorder, and to assess the type of nephropathy and its impact on renal function. In addition, it is mandatory to carefully search for extrarenal manifestations. To accurately characterize the renal disease, a kidney biopsy with detailed immunofluorescence (IF) and electron microscopic (EM) studies to identify deposit composition and pattern of organization is needed in most cases. The one exception is AL amyloidosis, which can be diagnosed if AL deposits can be demonstrated in other tissues such as fat.

Treatment of the B-cell clone

To date, no strategy is available to inhibit MIg tissue deposition or to directly clear the already deposited material. Innovative strategies that are currently in early clinical testing include small molecule weight inhibitors of serum amyloid P protein and monoclonal antibody against neoepitope on ALs.9  Thus, targeting the underlying B-cell clone with chemotherapy, although it is not an evidently malignant clone per se, is the only available therapeutic option for MGRS.

The choice of chemotherapeutic agents should take into account their renal metabolism and potential renal and extrarenal toxicity. For alkylating agents, cyclophosphamide is preferred to melphalan due to its lower toxicity in patients with reduced kidney function.10  Similarly, within the immunomodulatory drug class, thalidomide may be more appropriate than lenalidomide because the latter is, in part, cleared renally and may also sometimes worsen renal function in certain disease states notably in AL amyloidosis.11,12  In contrast, bortezomib can be used without dose adjustment with a good tolerance profile, including in the setting of ESRD.13-15  The risk of peripheral neuropathy remains a concern, but has been shown to be reduced by weekly administration and the use of the subcutaneous route of administration in patients with myeloma.16  Bendamustine, which has been proposed for the treatment of various lymphoid disorders, has also a predominantly nonrenal metabolism and can be given to patients with ESRD.17,18  Among nucleoside analogs, fludarabine requires dose adaptation and should be avoided in patients with renal failure.19  The use of rituximab and other anti-CD20 monoclonal antibodies raises no concerns in patients with renal impairment, including ESRD.20,21 

High-dose melphalan (HDM) supported by autologous peripheral blood stem cell transplantation (ASCT) may be a therapeutic option in some patients. It is essential to collect stem cells early in the course of the treatment, avoiding excessive prior use of drugs with potential stem cell damage, such as melphalan and lenalidomide. HDM/ASCT is feasible in multiple myeloma (MM) patients with renal failure, even requiring dialysis.22-24  However, mortality and morbidity, including the risk of worsening renal function, increases with the severity of renal impairment.25-27  Melphalan dose should be adjusted in patients with chronic kidney disease (CKD) stage 3 or above, and risk/benefit ratio should be carefully evaluated in each case.

The underlying plasma cell clone responsible for MGRS may manifest as a solitary plasmacytoma. Although this situation is rare, it has key therapeutic implications. Local radiotherapy may result in a complete and sustained control of the MIg production and, consequently, of its renal consequences.28  The initial work-up of MGRS should include a complete skeletal survey using conventional radiographs, and magnetic resonance imaging or positron emission tomography scan may be required for imaging suspected mass lesions or a solitary plasmacytoma.

In MGRS, assessment of hematological response is crucial. It depends on the evaluation of the MIg component that is responsible for renal lesions. In AL amyloidosis, measurement of serum free light chain (FLC) provides an essential tool. Current response criteria in AL amyloidosis distinguishes complete response (normal FLC ratio and negative serum and urine immunofixation), very good partial response (difference between involved and uninvolved FLCs <40 mg/L), partial response (difference between involved and uninvolved FLCs decrease >50%), and no response.29  The use of the same criteria in all types of MGRS due to the deposition of monoclonal light chain (LC), particularly light chain deposition disease (LCDD) and FS is logical. In MGRS involving an intact MIg, the criteria recommended in MM should be applied.30  When the causal MIg is not detectable or difficult to measure, evaluating the cellular response, usually by repeated bone marrow examination, is the only way to assess the hematologic efficacy of treatment. In all cases, renal (and extrarenal, if present) response should be regularly monitored, bearing in mind that it is usually delayed and depends on the quality of the hematologic response.31  Whether new molecular or cytometric techniques are useful to detect residual disease and impact outcomes deserves further investigation.32 

In all MGRS subtypes, evidence for relapse of the underlying clonal disease should prompt the clinician to reinitiate therapy based on criteria similar to the ones applied for the initiation of primary therapy. Treatment choice should take into account characteristics of the first response, toxicity of prior therapies, general status, and renal function.

Treatment of renal disease

MGRS should be monitored according to usual best practices including, for example, thrombotic and infectious risk prevention in case of nephrotic syndrome. Except in AL amyloidosis, hypertension and proteinuria should be controlled, preferably using blockers of the renin-angiotensin system. In patients with FS, bicarbonate, phosphate, and vitamin D supplementation should be given to prevent osteomalacia.33  For AL amyloidosis, introduction of an angiotensin-converting enzyme inhibitor and angiotensin receptor blocker should only be considered in patients who are hypertensive given these patients’ tendency toward orthostatic hypotension.

MGRS should not be considered as a contraindication to renal transplantation because the risk of patients dying from their clone is low. However, there are no data to suggest that small B-cell clones are truly curable, thus the risk of disease recurrence does expose the graft to risk of failure. This can occur after a variable delay depending on each type of renal disease. Recurrence of AL amyloidosis usually slowly impacts graft function, whereas graft loss rapidly occurs in most patients with PGNMID.33,34,34-39  Reducing the level of the MIg by obtaining the best hematologic response is a critical issue in allograft survival. Although this ideally means stringent complete response, a renal transplant may be considered in patients who are not in hematologic complete response and have no cardiac involvement.30,40  This must be discussed in each individual case, particularly taking into account the estimated time before the expected deterioration of renal function that could likely be irreversible. During posttransplant follow-up, careful surveillance of MIg parameters is mandatory. Reintroduction of therapy should be considered upon progression of the clonal disorder. Thus, the decision for renal transplantation should be taken considering the underlying MGRS characteristics, initial therapeutic response, presence of extrarenal manifestations, and the patient’s status.38  The risk of graft loss, its link with the B-cell clone and the potential need for reintroduction of chemotherapy should be clearly explained to the recipient (and to the donor, if a living donor transplantation is considered).

AL (AH and ALH) amyloidosis is usually associated with a low-grade plasma cell clone, most often secreting λ LC. The amyloid in AL is composed of monoclonal ALs, whereas it is composed of monoclonal AHs in AH, and ALH contains the entire immunoglobulin. Renal involvement is present in approximately 70% to 80% of patients and extrarenal manifestations are frequent.41-43  Approximately 75% of all AL patients present with proteinuria and 36% are in the nephrotic range. Elevated creatinine is noted in more than half the patients. Because the kidney is not an important contributor to early mortality in these patients, the type of treatment used is guided by the degree of cardiac involvement, as assessed by cardiac biomarkers. At the start of the 21st century, patients were now classified according to Mayo Clinic criteria as stage I, II, or III, depending on whether the N-terminal prohormone of brain natriuretic peptide and troponin T levels are both low (<332 ng/L and 0.035 µg/L), are high for only 1 level, or are both high, respectively.29,44  More recently, the level of immunoglobulin FLC was added to the criteria with minor modifications of cut-points and inclusion of FLC burden.45  Achieving the best and most durable hematologic response is the goal of therapy and must be adapted to patients who are often very fragile. The more severe the cardiac disease, the quicker response should be obtained. Because direct myocardial toxicity of amyloidogenic LC has been documented, rapid suppression of involved FLC is an important prognostic factor, particularly in patients with stage III cardiac disease.29,44,46-48 

Current recommendations can be summarized as follows:

  • In patients with stage I and II disease, first line treatment should be based on melphalan + dexamethasone (M-Dex). It is likely that reinforcing this regimen with bortezomib increases hematologic and organ response rate.49-52  This is currently under investigation through an international phase III trial comparing M-Dex with M-Dex plus bortezomib (clinical trial #NCT01277016). Until the results of this study are available, the current approach is to rapidly introduce bortezomib, after 1 or 2 courses of M-Dex in the absence of a clonal response. In patients with advanced CKD, cyclophosphamide is preferred to melphalan, and regimens such as cyclophosphamide-bortezomib-dexamethasone (CBD, also referred to as CyBorD or VCD) have demonstrated their efficacy.53,54  Another option is to use thalidomide instead of bortezomib (CTD regimen).55 

  • Patients with stage III cardiac involvement represent a therapeutic challenge because their median survival remains poor. Preliminary encouraging results have been obtained using the CBD regimen, which seems to significantly reduce the early death rate based on small case series.53,54  In young selected patients, cardiac transplantation can be considered, preferably after hematologic remission has been achieved.34,36 

  • In selected patients (mainly stage I and II disease), HDM/ASCT should be considered, in the absence of overt renal insufficiency and the absence of other advanced organ failure.56,57 

In MIDD, as with AL, the monoclonal gammopathy may occasionally be an overt myeloma (>10% bone marrow plasma cells with at least 1 myeloma-defining event); however, in MIDD, the clone more often secretes κ LC. Renal involvement is nearly constant and usually manifests with high-grade proteinuria, hematuria, hypertension, and renal insufficiency. Extrarenal manifestations, particularly cardiac and hepatic, are not uncommon, but are rarely symptomatic. The most frequent form of MIDD is LCDD. IF study of the kidney biopsy is mandatory for the diagnosis, showing typical linear amorphous LC deposits along the tubular basement membranes. Glomerular and vascular deposits are usually associated with a pattern of nodular glomerulosclerosis in two-thirds of patients with LCDD.58-61  Heavy chain deposition disease and light and heavy chain deposition disease are rare and should be managed similarly as LCDD.62,63 

As MIDD is rare, controlled studies are lacking and the therapeutic approach is based on consensus opinion. Achieving the best hematologic response appears to be as important as in AL. It may result in regression of MIg tissue deposits, providing that complete and sustained remission has been obtained.64,65 

Small retrospective series suggested that HDM/ASCT is a good therapeutic option, with high hematologic response rates and low treatment-related mortality. This contrasts with AL amyloidosis in which patients are much more fragile with frequent systemic complications that increase treatment-related mortality.64-71  Data regarding HDM/ASCT have been published before the era of novel antimyeloma agents. Preliminary results suggest that bortezomib-based regimens could produce hematologic response rates similar to those obtained with HDM/ASCT, as is the case in MM.72-77 

Taking into account these points, therapeutic recommendations should be based on the degree of renal impairment:

  • In patients with CKD stages 1 to 3, the main goal of treatment is preserving kidney function. The panel recommends the use of a bortezomib-based regimen, such as CBD, as front line. HDM/ASCT should then be considered in selected patients with good performance status and no significant extrarenal manifestations, particularly when they have achieved only partial hematological response to the initial treatment.

  • In patients with CKD stages 4 and 5, the probability of renal recovery is low. In patients not eligible for renal transplantation, the main goal of the treatment is preserving extrarenal organs, particularly the heart. The panel recommends a bortezomib-based regimen, such as CBD. If a renal transplantation is planned, the therapeutic goal is the preservation of long-term allograft function, which requires an optimal clonal response.59,78  Accordingly, HDM/ASCT should be considered after a 3 to 4 cycles of a CBD-like regimen.

Monoclonal immunoglobulin may precipitate under cold exposure, thus defining cryoglobulinemia. Type I cryoglobulins are composed of a single MIg.79  A serum rheumatoid factor (RF) is typically not detected and complement abnormalities are not constant. Type I cryoglobulinemia may be asymptomatic or cause cold-triggered ischemic symptoms, predominantly cutaneous (Raynaud’s phenomenon).80  Articular manifestations are mostly observed when the cryoglobulin (usually an IgG3) precipitates as crystals (crystal-cryoglobulinemia).81,82  Renal manifestations are more common with IgG type I cryoglobulin and less frequent with IgM.82,83  Cryoglobulinemia typically manifests as chronic glomerular disease with flares characterized by nephritic syndrome, acute renal insufficiency, and severe hypertension. Histologically, the hallmark of the disease is membranoproliferative glomerulonephritis with glomerular thrombi and microtubular deposits made up of the monoclonal cryoglobulin.82,83 

Type I cryoglobulinemia may be observed in patients who would otherwise be classified as monoclonal gammopathy of undetermined significance (MGUS) based on level of clonal burden, MM, Waldenström's macroglobulinemia (WM) or any other type of B-cell lymphoid disorder secreting an entire MIg.82-84  Patients should carefully avoid cold exposure and take appropriate protective measures. As data regarding management of this rare condition are scarce, the following recommendations are only guided by the panel experience:

  • In patients with few systemic symptoms and a low-grade underlying B-cell proliferation, observation alone is recommended, including serial assessment of renal parameters, because renal manifestations may occur secondarily.

  • In patients with symptomatic and/or progressive systemic disease, particularly in the presence of renal complications, therapy is indicated and should be selected based on the underlying clone:

    1. If it is plasmacytic, usually secreting a monoclonal IgG (or IgA), treatment should rely on antimyeloma agents. In patients with renal failure, bortezomib, cyclophosphamide, and/or thalidomide-based regimens should be used. Rituximab is not indicated. In selected patients, HDM/ASCT may be considered.

    2. If it is lymphoplasmacytic, usually associated with a monoclonal IgM, the treatment should be that of WM, currently based on Rituximab-containing regimens.

    3. If the underlying disease is chronic lymphocytic leukemia (CLL) or a B-cell lymphoma of any type, then the treatment should be adapted accordingly.

In all cases, bendamustine, which is not eliminated by the kidney, is likely to be a good alternative. In patients with acute severe systemic symptoms, complete plasma exchange is indicated in addition to chemotherapy.82-84 

Type II mixed cryoglobulins are composed of an MIg, usually an IgM κ with a RF activity, associated with polyclonal immunoglobulin. A serum RF is always detectable and serum complement levels are constantly decreased. Type II cryoglobulinemia is often a silent condition. When symptomatic, vascular purpuric lesions are nearly always present. Other manifestations are usually related to small vessel vasculitis, including arthralgias and/or peripheral neuropathy. Disease flares are common and more often triggered by activity and orthostatism than by cold exposure.78,79,84  Clinical and pathological renal manifestations are close to that observed in type I cryoglobulinemia. However, IF studies show that glomerular deposits contain both the monoclonal IgM and polyclonal IgG along with complement components.81 

Most cases are associated with chronic hepatitis C virus (HCV) infection.85  Other conditions include hepatitis B virus infection and autoimmune disorders, particularly Sjögren’s syndrome. In all cases, type II cryoglobulinemia implicates the presence of an underlying B-cell clone most typically a very small clone with less than 10% bone marrow involvement. An overt lymphoid proliferation, usually a WM or a low-grade lymphoma, can be detected initially or may develop during follow-up.79,86 

The knowledge of whether or not type II cryoglobulinemia is associated with HCV infection is imperative to guide treatment. Accordingly, HCV testing infection should be systematic, including serum HCV RNA detection, and genotyping if present.85,87 

In the absence of prospective studies, treatment strategy may be summarized as follows:

  • Antiviral therapy, usually combining pegylated α interferon and ribavirin should be given to all patients with symptomatic type II cryoglobulinemia associated with chronic HCV infection. Antiviral therapy may be given alone in patients who have only a few symptoms (ie, episodic flares of vasculitic purpura). In those with more symptomatic vasculitis, antiviral therapy should be combined with rituximab. In addition, in patients with rapidly progressive renal disease and/or other severe organ involvement, total plasma exchange should be considered. Importantly because of their remarkable symptomatic efficacy, high-dose steroids should be added in all cases with overt vasculitis. Due to the risk of their side effects, one should avoid long-term administration of steroids if possible.88,89 

  • In those patients with no detectable viral replication, who present with episodic purpuric flares, surveillance only is recommended. In case of recurrent symptoms or of renal involvement, rituximab is the treatment of choice.90 

  • In all patients with overt WM or B-cell lymphoma, chemotherapy should be considered in patients with symptoms more significant than occasional purpura, regardless of their HCV status. The regimen should be defined by taking into account the type of the underlying B-cell clone and the level of renal function (see previously). Rituximab-containing regimens and bendamustine can be used in all levels of renal function.91 

ITG, also referred to as glomerulonephritis with organized microtubular monoclonal immunoglobulin deposits is a rare glomerular disease with proteinuria, frequent nephrotic syndrome, hematuria, CKD, and hypertension. Extrarenal manifestations are uncommon. Glomerular lesions are usually characterized by atypical membranous and MPGN patterns. By IF, glomerular deposits are composed of monotypic IgG and complement components. EM, which is required for the diagnosis, shows a typical organization of deposits into microtubules of 10 to 60 nm in diameter, arranged in parallel bundles.3,5,6,92  Similar microtubular inclusions may be observed in the cytoplasm of the circulating and medullary clonal B-cell population.3,5 

Importantly, the underlying B-cell lymphoid disorder is a CLL or a small lymphocytic lymphoma in more than half of the cases. A low-grade plasma cell clone is less frequent.5,92  Accordingly, the initial workup should include a careful search for a clonal B-cell population among peripheral blood and bone marrow lymphocytes, including phenotypic and immunoglobulin gene rearrangement studies.

Although therapeutic choice relies on a few, small case series, a CLL-adapted treatment may be proposed in most patients. In case of severe CKD, cyclophosphamide and/or bendamustine-based regimens, including corticosteroids, may be recommended. The addition of rituximab should be considered in patients with overt CLL. In patients with gammopathy only, the role of rituximab is questionable and bortezomib-based therapy may be considered.5,92 

PGNMID is a recently described entity that should be distinguished from Randall-type MIDD. Both diseases are featured by glomerular nonorganized MIg deposits. However, in PGNMID, deposits usually consist of an entire MIg (most commonly IgG3κ), with a granular nonlinear appearance by IF and EM, without detectable deposits along tubular basement membranes and around vascular myocytes. Various patterns can be observed, including mesangial, atypical membranous, and membranoproliferative glomerulonephritis. Globally, PGNMID resembles an immune complex-like glomerulonephritis, but with monotypic immunoglobulin deposits, a feature that should draw the pathologist’s attention.93-96 

Extrarenal manifestations are rare and PGNMID appears as a renal limited disorder with prominent glomerular symptoms and frequent CKD. Importantly, sensitive techniques including nephelometric tests for FLCs can detect a serum and urine MIg in approximately one-third of patients only. A monoclonal proliferation of plasma cells in the bone marrow is found in less than 10% of patients.93-96 

Some authors are reluctant to recommend chemotherapy in PGNMID with no detectable MIg. However, monotypic glomerular deposits are the result of a circulating MIg, which indicates the presence of an underlying B-cell disorder. Consequently, PGNMID almost always recurs after renal transplantation.37,97 

The panel recommends adapting the therapeutic approach to the severity of renal disease:

  • In patients with stages 1 and 2 CKD, proteinuria of less than 1 g/day and no evidence of progressive disease, symptomatic measures only may be advised with careful surveillance. In such cases, occasional spontaneous renal remissions may occur.

  • In patients with stages 1 and 2 CKD and high-grade proteinuria (>1 g/day) or progressive disease, and in patients with stages 3 and 4 CKD, chemotherapy is indicated. Cyclophosphamide and bortezomib are the drugs of choice, and a CBD-like regimen is a good option. In some patients aged <65 years, HDM/ASCT may be performed. Preliminary reports suggest a beneficial effect of rituximab, including the effect in patients without a detectable B-cell clone.90  However, it seems reasonable to propose rituximab only to patients in whom an associated CD-20 positive B-cell clone can be demonstrated.

  • In patients with stage 5 CKD who are candidates for renal transplantation, the achievement of a complete hematologic remission is a key goal for patients with a detectable monoclonal gammopathy.37,97  Thus, HDM/ ASCT should be strongly considered. For those who never had a detectable monoclonal gammopathy or plasma cell clone, there is no consensus regarding their treatment prior to kidney transplant. In contrast, in patients ineligible for renal transplantation, the benefit of chemotherapy is highly questionable and conservative treatment should be recommended.

Acquired FS is characterized by proximal tubular dysfunction secondary to accumulation of monoclonal κ LCs crystalline inclusions within the endolysosomal compartment of proximal tubular cells. Similar LC inclusions can be detected in the cytoplasm of associated clonal plasma cells. FS typically complicates MGUS or a low-grade MM, which is almost always of the κ class.4,33,98,99  Few cases have been described in WM.100  Most patients present with moderate CKD, with unusual hypophosphatemia and hypouricemia, which should prompt the search for other signs of proximal tubule dysfunction, particularly generalized aminoaciduria. Of note, the urine phosphate leak frequently causes osteomalacia and subsequent bone pain that should not be misinterpreted as secondary to myeloma.

Other extrarenal manifestations are absent in typical FS. However, FS may be part of the so-called crystal-storing histiocytosis (CSH), which is characterized by the accumulation of LC crystals, not only in proximal tubular cells, but also in lysosomes of histiocytes of bone marrow. In addition, CSH can involve various tissues, including spleen and lymph nodes, sometimes in the context of systemic symptoms with macrophage activation.101,102 

Very few series of LC-associated FS have been published, and the efficacy of the so-called novel anti-myeloma agents has not been evaluated. In most cases, FS appears to slowly progress toward ESRD and rarely symptomatic myeloma.33,98  Accordingly, therapeutic decisions should take into account treatment side effects, particularly the potential risk of secondary myelodysplastic syndrome from alkylating agents.98 

Symptomatic measures to prevent osteomalacia are mandatory. All patients with an associated overt lymphoid disorder should receive appropriate chemotherapy. For the rare patients with symptomatic CSH, steroids should be considered in addition to chemotherapy.

Otherwise, treatment choices should be adapted to the degree of renal failure:

  • In patients with stages 1 to 3 CKD, chemotherapy should be considered to try to slow progression to ESRD. Cyclophosphamide-, bortezomib-, or thalidomide-based regimens are the best options. Bendamustine may also be used. HDM/ASCT may be performed in selected nonresponding patients, although the benefit of this strategy remains to be proven.

  • In patients with stages 4 to 5 CKD who are eligible for renal allograft, chemotherapy, including HDM/ASCT, should be considered either prior to and/or after transplantation. In patients who will not be candidates for renal transplantation, there is no benefit to introduce chemotherapy.

In addition to kidney deposition of a MIg, other mechanisms may induce renal lesions in MGRS. They may involve the secretion of various biological factors and/or autoantibody activity of the MIg.

The so-called POEMS (Polyneuropathy, Organomegaly, Endocrinopathy, M-protein, and Skin abnormalities) syndrome can include renal manifestations, usually featured by vascular and glomerular thrombotic microangiopathy including mesangiolysis. These are considered to be due to the secretion of vascular endothelial growth factor by the clonal cells or their environment, which is a hallmark of this syndrome. Whether the causal plasmacytic proliferation is localized or not is the key point for therapeutic decisions.103 

A novel MGRS entity characterized by glomerulonephritis with isolated glomerular C3 deposits has been recently described. It is associated with a circulating monoclonal IgG, most often with MGUS or indolent MM. Hypocomplementemia secondary to activation of the complement alternative pathway is usual, in the absence of detectable anti-C3 convertase activity (nephritic factor). Autoantibody activity of the MIg against a complement alternative pathway regulatory protein is the main current hypothesis. Because the disease course is rapid, with a high risk of recurrence after renal transplantation, chemotherapy should be given early.104,105 

Current treatment of MGRS is based on therapies targeting the causal B-cell clone with treatment choices based on extrapolation of treatments used for the equivalent overt malignancy. Therapeutic choices should take into account the renal characteristics of the disease, particularly the risk of CKD progression, the presence and severity of extrarenal manifestations, and the safety profile of antineoplastic drugs in renal impairment. Early diagnosis, when renal function is still preserved, usually facilitates treatment management and results in better long-term outcome. Because MGRS is a heterogeneous and relatively rare entity, a common effort of both nephrologists and hematologists inside well-designed prospective collaborative studies is required to improve management.

Contribution: J.-P.F., F.B., R.A.K., E.K., B.M.W., M.A.C., M.T.D., A.D., and N.L. all contributed to the planning and writing of this manuscript.

Conflict-of-interest disclosure: A.D. received research funding from Janssen, Pfizer, Millennium, and Celgene; M.A.C. received honoraria from Celgene and Janssen, and research funding from Celgene; and B.W. received research funding from Janssen. The remaining authors declare no competing financial interests.

Correspondence: Jean-Paul Fermand, Immuno-Hematology Unit, Saint-Louis Hospital, Paris, France; e-mail: jpfermand@yahoo.fr; and Nelson Leung, Division of Nephrology and Hypertension and Division of Hematology, Mayo Clinic, Rochester, MN; e-mail: leung.nelson@mayo.edu.

1
Leung
 
N
Bridoux
 
F
Hutchison
 
CA
, et al. 
International Kidney and Monoclonal Gammopathy Research Group. Monoclonal gammopathy of renal significance: when MGUS is no longer undetermined or insignificant.
Blood
2012
, vol. 
120
 
22
(pg. 
4292
-
4295
)
2
Merlini
 
G
Stone
 
MJ
Dangerous small B-cell clones.
Blood
2006
, vol. 
108
 
8
(pg. 
2520
-
2530
)
3
Touchard
 
G
Touchard
 
G
Aucouturier
 
P
Hermine
 
O
Ronco
 
P
Ultrastructural pattern and classification of renal monoclonal immunoglobulin deposits.
Monoclonal Gammopathies and the Kidney
2003
Dordrecht, Boston
Kluwer Academic Publishers
(pg. 
95
-
118
)
4
Ronco
 
P
Bridoux
 
F
Aucouturier
 
P
Coffman
 
TM
Falk
 
RJ
Molitoris
 
BA
Neilson
 
EG
Schrier
 
RW
Monoclonal Gammopathies: Multiple Myeloma, Amyloidosis, and Related Disorders.
Schrier's Diseases of the Kidney 9th ed
2013
Philadelphia: Wolters Kluwer/Lippincott Williams & Wilkins
(pg. 
1710
-
1765
)
5
Bridoux
 
F
Hugue
 
V
Coldefy
 
O
, et al. 
Fibrillary glomerulonephritis and immunotactoid (microtubular) glomerulopathy are associated with distinct immunologic features.
Kidney Int
2002
, vol. 
62
 
5
(pg. 
1764
-
1775
)
6
Rosenstock
 
JL
Markowitz
 
GS
Valeri
 
AM
Sacchi
 
G
Appel
 
GB
D’Agati
 
VD
Fibrillary and immunotactoid glomerulonephritis: Distinct entities with different clinical and pathologic features.
Kidney Int
2003
, vol. 
63
 
4
(pg. 
1450
-
1461
)
7
Gallo
 
G
Picken
 
M
Buxbaum
 
J
Frangione
 
B
The spectrum of monoclonal immunoglobulin deposition disease associated with immunocytic dyscrasias.
Semin Hematol
1989
, vol. 
26
 
3
(pg. 
234
-
245
)
8
Lebovic
 
D
Hoffman
 
J
Levine
 
BM
, et al. 
Predictors of survival in patients with systemic light-chain amyloidosis and cardiac involvement initially ineligible for stem cell transplantation and treated with oral melphalan and dexamethasone.
Br J Haematol
2008
, vol. 
143
 
3
(pg. 
369
-
373
)
9
Bodin
 
K
Ellmerich
 
S
Kahan
 
MC
, et al. 
Antibodies to human serum amyloid P component eliminate visceral amyloid deposits.
Nature
2010
, vol. 
468
 
7320
(pg. 
93
-
97
)
10
Carlson
 
K
Hjorth
 
M
Knudsen
 
LM
Nordic Myeloma Study Group
Toxicity in standard melphalan-prednisone therapy among myeloma patients with renal failure—a retrospective analysis and recommendations for dose adjustment.
Br J Haematol
2005
, vol. 
128
 
5
(pg. 
631
-
635
)
11
Chen
 
N
Lau
 
H
Kong
 
L
Kumar
 
G
Zeldis
 
JB
Knight
 
R
Laskin
 
OL
Pharmacokinetics of lenalidomide in subjects with various degrees of renal impairment and in subjects on hemodialysis.
J Clin Pharmacol
2007
, vol. 
47
 
12
(pg. 
1466
-
1475
)
12
Specter
 
R
Sanchorawala
 
V
Seldin
 
DC
, et al. 
Kidney dysfunction during lenalidomide treatment for AL amyloidosis.
Nephrol Dial Transplant
2011
, vol. 
26
 
3
(pg. 
881
-
886
)
13
Jagannath
 
S
Barlogie
 
B
Berenson
 
JR
, et al. 
SUMMIT/CREST Investigators
Bortezomib in recurrent and/or refractory multiple myeloma. Initial clinical experience in patients with impared renal function.
Cancer
2005
, vol. 
103
 
6
(pg. 
1195
-
1200
)
14
Chanan-Khan
 
AA
Kaufman
 
JL
Mehta
 
J
, et al. 
Activity and safety of bortezomib in multiple myeloma patients with advanced renal failure: a multicenter retrospective study.
Blood
2007
, vol. 
109
 
6
(pg. 
2604
-
2606
)
15
San-Miguel
 
JF
Richardson
 
PG
Sonneveld
 
P
, et al. 
Efficacy and safety of bortezomib in patients with renal impairment: results from the APEX phase 3 study.
Leukemia
2008
, vol. 
22
 
4
(pg. 
842
-
849
)
16
Moreau
 
P
Pylypenko
 
H
Grosicki
 
S
, et al. 
Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study.
Lancet Oncol
2011
, vol. 
12
 
5
(pg. 
431
-
440
)
17
Ramasamy
 
K
Hazel
 
B
Mahmood
 
S
Corderoy
 
S
Schey
 
S
Bendamustine in combination with thalidomide and dexamethasone is an effective therapy for myeloma patients with end stage renal disease.
Br J Haematol
2011
, vol. 
155
 
5
(pg. 
632
-
634
)
18
Pönisch
 
W
Andrea
 
M
Wagner
 
I
, et al. 
Successful treatment of patients with newly diagnosed/untreated multiple myeloma and advanced renal failure using bortezomib in combination with bendamustine and prednisone.
J Cancer Res Clin Oncol
2012
, vol. 
138
 
8
(pg. 
1405
-
1412
)
19
Lichtman
 
SM
Etcubanas
 
E
Budman
 
DR
, et al. 
The pharmacokinetics and pharmacodynamics of fludarabine phosphate in patients with renal impairment: a prospective dose adjustment study.
Cancer Invest
2002
, vol. 
20
 
7-8
(pg. 
904
-
913
)
20
Jillella
 
AP
Dainer
 
PM
Kallab
 
AM
Ustun
 
C
Treatment of a patient with end-stage renal disease with Rituximab: pharmacokinetic evaluation suggests Rituximab is not eliminated by hemodialysis.
Am J Hematol
2002
, vol. 
71
 
3
(pg. 
219
-
222
)
21
Vieira
 
CA
Agarwal
 
A
Book
 
BK
, et al. 
Rituximab for reduction of anti-HLA antibodies in patients awaiting renal transplantation: 1. Safety, pharmacodynamics, and pharmacokinetics.
Transplantation
2004
, vol. 
77
 
4
(pg. 
542
-
548
)
22
San Miguel
 
JF
Lahuerta
 
JJ
Garcia-Sanz
 
R
, et al. 
Are myeloma patients with renal failure candidates for autologous stem cell transplantation?
Hematol J
2000
, vol. 
1
 
1
(pg. 
28
-
36
)
23
Badros
 
A
Barlogie
 
B
Siegel
 
E
, et al. 
Results of autologous stem cell transplant in multiple myeloma patients with renal failure.
Br J Haematol
2001
, vol. 
114
 
4
(pg. 
822
-
829
)
24
Knudsen
 
LM
Nielsen
 
B
Gimsing
 
P
Geisler
 
C
Autologous stem cell transplantation in multiple myeloma: outcome in patients with renal failure.
Eur J Haematol
2005
, vol. 
75
 
1
(pg. 
27
-
33
)
25
Schrier
 
RW
Parikh
 
CR
Comparison of renal injury in myeloablative autologous, myeloablative allogeneic and non-myeloablative allogeneic haematopoietic cell transplantation.
Nephrol Dial Transplant
2005
, vol. 
20
 
4
(pg. 
678
-
683
)
26
Ando
 
M
Mori
 
J
Ohashi
 
K
, et al. 
A comparative assessment of the RIFLE, AKIN and conventional criteria for acute kidney injury after hematopoietic SCT.
Bone Marrow Transplant
2010
, vol. 
45
 
9
(pg. 
1427
-
1434
)
27
Irazabal
 
MV
Eirin
 
A
Gertz
 
MA
, et al. 
Acute kidney injury during leukocyte engraftment after autologous stem cell transplantation in patients with light-chain amyloidosis.
Am J Hematol
2012
, vol. 
87
 
1
(pg. 
51
-
54
)
28
Dingli
 
D
Kyle
 
RA
Rajkumar
 
SV
, et al. 
Immunoglobulin free light chains and solitary plasmacytoma of bone.
Blood
2006
, vol. 
108
 
6
(pg. 
1979
-
1983
)
29
Palladini
 
G
Dispenzieri
 
A
Gertz
 
MA
, et al. 
New criteria for response to treatment in immunoglobulin light chain amyloidosis based on free light chain measurement and cardiac biomarkers: impact on survival outcomes.
J Clin Oncol
2012
, vol. 
30
 
36
(pg. 
4541
-
4549
)
30
Durie
 
BG
Harousseau
 
JL
Miguel
 
JS
, et al. 
International uniform response criteria for multiple myeloma.
Leukemia
2006
, vol. 
20
 
12
(pg. 
1467
-
1673
)
31
Leung
 
N
Dispenzieri
 
A
Fervenza
 
FC
, et al. 
Renal response after high-dose melphalan and stem cell transplantation is a favorable marker in patients with primary systemic amyloidosis.
Am J Kidney Dis
2005
, vol. 
46
 
2
(pg. 
270
-
277
)
32
Ramirez-Alvarado
 
M
Ward
 
CJ
Huang
 
BQ
, et al. 
Differences in immunoglobulin light chain species found in urinary exosomes in light chain amyloidosis (Al).
PLoS ONE
2012
, vol. 
7
 
6
pg. 
e38061
 
33
Messiaen
 
T
Deret
 
S
Mougenot
 
B
, et al. 
Adult Fanconi syndrome secondary to light chain gammopathy. Clinicopathologic heterogeneity and unusual features in 11 patients.
Medicine (Baltimore)
2000
, vol. 
79
 
3
(pg. 
135
-
154
)
34
Sattianayagam
 
PT
Gibbs
 
SD
Pinney
 
JH
, et al. 
Solid organ transplantation in AL amyloidosis.
Am J Transplant
2010
, vol. 
10
 
9
(pg. 
2124
-
2131
)
35
Herrmann
 
SM
Gertz
 
MA
Stegall
 
MD
, et al. 
Long-term outcomes of patients with light chain amyloidosis (AL) after renal transplantation with or without stem cell transplantation.
Nephrol Dial Transplant
2011
, vol. 
26
 
6
(pg. 
2032
-
2036
)
36
Mignot
 
A
Varnous
 
S
Redonnet
 
M
, et al. 
Heart transplantation in systemic (AL) amyloidosis: a retrospective study of eight French patients.
Arch Cardiovasc Dis
2008
, vol. 
101
 
9
(pg. 
523
-
532
)
37
Nasr
 
SH
Sethi
 
S
Cornell
 
LD
, et al. 
Proliferative glomerulonephritis with monoclonal IgG deposits recurs in the allograft.
Clin J Am Soc Nephrol
2011
, vol. 
6
 
1
(pg. 
122
-
132
)
38
Bridoux
 
F
Ronco
 
P
Gillmore
 
J
Fermand
 
JP
Renal transplantation in light chain amyloidosis: coming out of the cupboard.
Nephrol Dial Transplant
2011
, vol. 
26
 
6
(pg. 
1766
-
1768
)
39
Pinney
 
JH
Lachmann
 
HJ
Sattianayagam
 
PT
, et al. 
Renal transplantation in systemic amyloidosis-importance of amyloid fibril type and precursor protein abundance.
Am J Transplant
2013
, vol. 
13
 
2
(pg. 
433
-
441
)
40
Leung
 
N
Griffin
 
MD
Dispenzieri
 
A
, et al. 
Living donor kidney and autologous stem cell transplantation for primary systemic amyloidosis (AL) with predominant renal involvement.
Am J Transplant
2005
, vol. 
5
 
7
(pg. 
1660
-
1670
)
41
Kyle
 
RA
Gertz
 
MA
Primary systemic amyloidosis: clinical and laboratory features in 474 cases.
Semin Hematol
1995
, vol. 
32
 
1
(pg. 
45
-
59
)
42
Kyle
 
RA
Linos
 
A
Beard
 
CM
Linke
 
RP
Gertz
 
MA
O’Fallon
 
WM
Kurland
 
LT
Incidence and natural history of primary systemic amyloidosis in Olmsted County, Minnesota, 1950 through 1989.
Blood
1992
, vol. 
79
 
7
(pg. 
1817
-
1822
)
43
Pinney
 
JH
Lachmann
 
HJ
Bansi
 
L
, et al. 
Outcome in renal AL amyloidosis after chemotherapy.
J Clin Oncol
2011
, vol. 
29
 
6
(pg. 
674
-
681
)
44
Dispenzieri
 
A
Gertz
 
MA
Kyle
 
RA
, et al. 
Serum cardiac troponins and N-terminal pro-brain natriuretic peptide: a staging system for primary systemic amyloidosis.
J Clin Oncol
2004
, vol. 
22
 
18
(pg. 
3751
-
3757
)
45
Kumar
 
S
Dispenzieri
 
A
Lacy
 
MQ
, et al. 
Revised prognostic staging system for light chain amyloidosis incorporating cardiac biomarkers and serum free light chain measurements.
J Clin Oncol
2012
, vol. 
30
 
9
(pg. 
989
-
995
)
46
Brenner
 
DA
Jain
 
M
Pimentel
 
DR
, et al. 
Human amyloidogenic light chains directly impair cardiomyocyte function through an increase in cellular oxidant stress.
Circ Res
2004
, vol. 
94
 
8
(pg. 
1008
-
1010
)
47
Shi
 
J
Guan
 
J
Jiang
 
B
, et al. 
Amyloidogenic light chains induce cardiomyocyte contractile dysfunction and apoptosis via a non-canonical p38alpha MAPK pathway.
Proc Natl Acad Sci USA
2010
, vol. 
107
 
9
(pg. 
4188
-
4193
)
48
Palladini
 
G
Barassi
 
A
Klersy
 
C
, et al. 
The combination of high-sensitivity cardiac troponin T (hs-cTnT) at presentation and changes in N-terminal natriuretic peptide type B (NT-proBNP) after chemotherapy best predicts survival in AL amyloidosis.
Blood
2010
, vol. 
116
 
18
(pg. 
3426
-
3430
)
49
Wechalekar
 
AD
Lachmann
 
HJ
Offer
 
M
Hawkins
 
PN
Gillmore
 
JD
Efficacy of bortezomib in systemic AL amyloidosis with relapsed/refractory clonal disease.
Haematologica
2008
, vol. 
93
 
2
(pg. 
295
-
298
)
50
Reece
 
DE
Sanchorawala
 
V
Hegenbart
 
U
, et al. 
VELCADE CAN2007 Study Group
Weekly and twice-weekly bortezomib in patients with systemic AL amyloidosis: results of a phase 1 dose-escalation study.
Blood
2009
, vol. 
114
 
8
(pg. 
1489
-
1497
)
51
Kastritis
 
E
Wechalekar
 
AD
Dimopoulos
 
MA
, et al. 
Bortezomib with or without dexamethasone in primary systemic (light chain) amyloidosis.
J Clin Oncol
2010
, vol. 
28
 
6
(pg. 
1031
-
1037
)
52
Reece
 
DE
Hegenbart
 
U
Sanchorawala
 
V
, et al. 
Efficacy and safety of once-weekly and twice-weekly bortezomib in patients with relapsed systemic AL amyloidosis: results of a phase 1/2 study.
Blood
2011
, vol. 
118
 
4
(pg. 
865
-
873
)
53
Venner
 
CP
Lane
 
T
Foard
 
D
, et al. 
Cyclophosphamide, bortezomib, and dexamethasone therapy in AL amyloidosis is associated with high clonal response rates and prolonged progression-free survival.
Blood
2012
, vol. 
119
 
19
(pg. 
4387
-
4390
)
54
Mikhael
 
JR
Schuster
 
SR
Jimenez-Zepeda
 
VH
, et al. 
Cyclophosphamide-bortezomib-dexamethasone (CyBorD) produces rapid and complete hematologic response in patients with AL amyloidosis.
Blood
2012
, vol. 
119
 
19
(pg. 
4391
-
4394
)
55
Wechalekar
 
AD
Goodman
 
HJ
Lachmann
 
HJ
Offer
 
M
Hawkins
 
PN
Gillmore
 
JD
Safety and efficacy of risk-adapted cyclophosphamide, thalidomide, and dexamethasone in systemic AL amyloidosis.
Blood
2007
, vol. 
109
 
2
(pg. 
457
-
464
)
56
Cibeira
 
MT
Sanchorawala
 
V
Seldin
 
DC
, et al. 
Outcome of AL amyloidosis after high-dose melphalan and autologous stem cell transplantation: long-term results in a series of 421 patients.
Blood
2011
, vol. 
118
 
16
(pg. 
4346
-
4352
)
57
Cordes
 
S
Dispenzieri
 
A
Lacy
 
MQ
, et al. 
Ten-year survival after autologous stem cell transplantation for immunoglobulin light chain amyloidosis.
Cancer
2012
, vol. 
118
 
24
(pg. 
6105
-
6109
)
58
Ronco
 
P
Plaisier
 
E
Mougenot
 
B
Aucouturier
 
P
Immunoglobulin light (heavy)-chain deposition disease: from molecular medicine to pathophysiology-driven therapy.
Clin J Am Soc Nephrol
2006
, vol. 
1
 
6
(pg. 
1342
-
1350
)
59
Nasr
 
SH
Valeri
 
AM
Cornell
 
LD
Fidler
 
ME
Sethi
 
S
D’Agati
 
VD
Leung
 
N
Renal monoclonal immunoglobulin deposition disease: a report of 64 patients from a single institution.
Clin J Am Soc Nephrol
2012
, vol. 
7
 
2
(pg. 
231
-
239
)
60
Lin
 
J
Markowitz
 
GS
Valeri
 
AM
Kambham
 
N
Sherman
 
WH
Appel
 
GB
D’Agati
 
VD
Renal monoclonal immunoglobulin deposition disease: the disease spectrum.
J Am Soc Nephrol
2001
, vol. 
12
 
7
(pg. 
1482
-
1492
)
61
Pozzi
 
C
D'Amico
 
Fogazzi
 
GB
, et al. 
Light chain deposition disease with renal involvement: clinical characteristics and prognostic factors.
Am J Kidney Dis
2003
, vol. 
42
 
6
(pg. 
1154
-
1163
)
62
Aucouturier
 
P
Khamlichi
 
AA
Touchard
 
G
, et al. 
Brief report: heavy-chain deposition disease.
N Engl J Med
1993
, vol. 
329
 
19
(pg. 
1389
-
1393
)
63
Moulin
 
B
Deret
 
S
Mariette
 
X
, et al. 
Nodular glomerulosclerosis with deposition of monoclonal immunoglobulin heavy chains lacking C(H)1.
J Am Soc Nephrol
1999
, vol. 
10
 
3
(pg. 
519
-
528
)
64
Royer
 
B
Arnulf
 
B
Martinez
 
F
, et al. 
High dose chemotherapy in light chain or light and heavy chain deposition disease.
Kidney Int
2004
, vol. 
65
 
2
(pg. 
642
-
648
)
65
Harada
 
K
Akai
 
Y
Sakan
 
H
Yamaguchi
 
Y
Nakatani
 
K
Iwano
 
M
Saito
 
Y
Resolution of mesangial light chain deposits 3 years after high-dose melphalan with autologous peripheral blood stem cell transplantation.
Clin Nephrol
2010
, vol. 
74
 
5
(pg. 
384
-
388
)
66
Firkin
 
F
Hill
 
PA
Dwyer
 
K
Gock
 
H
Reversal of dialysis-dependent renal failure in light-chain deposition disease by autologous peripheral blood stem cell transplantation.
Am J Kidney Dis
2004
, vol. 
44
 
3
(pg. 
551
-
555
)
67
Weichman
 
K
Dember
 
LM
Prokaeva
 
T
, et al. 
Clinical and molecular characteristics of patients with non-amyloid light chain deposition disorders, and outcome following treatment with high-dose melphalan and autologous stem cell transplantation.
Bone Marrow Transplant
2006
, vol. 
38
 
5
(pg. 
339
-
343
)
68
Lorenz
 
EC
Gertz
 
MA
Fervenza
 
FC
, et al. 
Long-term outcome of autologous stem cell transplantation in light chain deposition disease.
Nephrol Dial Transplant
2008
, vol. 
23
 
6
(pg. 
2052
-
2057
)
69
Hassoun
 
H
Flombaum
 
C
D’Agati
 
VD
, et al. 
High-dose melphalan and auto-SCT in patients with monoclonal Ig deposition disease.
Bone Marrow Transplant
2008
, vol. 
42
 
6
(pg. 
405
-
412
)
70
Telio
 
D
Shepherd
 
J
Forrest
 
D
Zypchen
 
L
Barnett
 
M
Nevill
 
T
Song
 
KW
High-dose melphalan followed by auto-SCT has favorable safety and efficacy in selected patients with light chain deposition disease and light and heavy chain deposition disease.
Bone Marrow Transplant
2012
, vol. 
47
 
3
(pg. 
453
-
455
)
71
Brioli
 
A
Zamagni
 
E
Pasquali
 
S
, et al. 
Long-term follow-up after autologous stem cell transplantation for light- and heavy-chain deposition disease.
Bone Marrow Transplant
2012
, vol. 
47
 
9
(pg. 
1248
-
1249
)
72
Kastritis
 
E
Migkou
 
M
Gavriatopoulou
 
M
Zirogiannis
 
P
Hadjikonstantinou
 
V
Dimopoulos
 
MA
Treatment of light chain deposition disease with bortezomib and dexamethasone.
Haematologica
2009
, vol. 
94
 
2
(pg. 
300
-
302
)
73
Kaposztas
 
Z
Kahan
 
BD
Katz
 
SM
Van Buren
 
CT
Cherem
 
L
Bortezomib successfully reverses early recurrence of light-chain deposition disease in a renal allograft: a case report.
Transplant Proc
2009
, vol. 
41
 
10
(pg. 
4407
-
4410
)
74
Gharwan
 
H
Truica
 
CI
Bortezomib-based chemotherapy for light chain deposition disease presenting as acute renal failure.
Med Oncol
2012
, vol. 
29
 
2
(pg. 
1197
-
1201
)
75
Tovar
 
N
Cibeira
 
MT
Rosiñol
 
L
, et al. 
Bortezomib/dexamethasone followed by autologous stem cell transplantation as front line treatment for light-chain deposition disease.
Eur J Haematol
2012
, vol. 
89
 
4
(pg. 
340
-
344
)
76
Jimenez-Zepeda
 
VH
Trudel
 
S
Winter
 
A
Reece
 
DE
Chen
 
C
Kukreti
 
V
Autologous stem cell transplant for light chain deposition disease: incorporating bortezomib to the induction therapy.
Am J Hematol
2012
, vol. 
87
 
8
(pg. 
822
-
823
)
77
Stewart
 
AK
Richardson
 
PG
San-Miguel
 
JF
How I treat multiple myeloma in younger patients.
Blood
2009
, vol. 
114
 
27
(pg. 
5436
-
5443
)
78
Leung
 
N
Lager
 
DJ
Gertz
 
MA
Wilson
 
K
Kanakiriya
 
S
Fervenza
 
FC
Long-term outcome of renal transplantation in light-chain deposition disease.
Am J Kidney Dis
2004
, vol. 
43
 
1
(pg. 
147
-
153
)
79
Brouet
 
JC
Clauvel
 
JP
Danon
 
F
Klein
 
M
Seligmann
 
M
Biologic and clinical significance of cryoglobulins. A report of 86 cases.
Am J Med
1974
, vol. 
57
 
5
(pg. 
775
-
788
)
80
Trejo
 
O
Ramos-Casals
 
M
García-Carrasco
 
M
, et al. 
Cryoglobulinemia: study of etiologic factors and clinical and immunologic features in 443 patients from a single center.
Medicine (Baltimore)
2001
, vol. 
80
 
4
(pg. 
252
-
262
)
81
Papo
 
T
Musset
 
L
Bardin
 
T
, et al. 
Cryocrystalglobulinemia as a cause of systemic vasculopathy and widespread erosive arthropathy.
Arthritis Rheum
1996
, vol. 
39
 
2
(pg. 
335
-
340
)
82
Bridoux
 
F
Sirac
 
C
Jaccard
 
A
, et al. 
Mason
 
JC
Pusey
 
CD
Renal Disease in Cryoglobulinemic Vasculitis.
The kidney in systemic autoimmune diseases
 
Amsterdam, Boston: Elsevier;2008:215-240
83
Karras
 
A
Noël
 
LH
Droz
 
D
, et al. 
Renal involvement in monoclonal (type I) cryoglobulinemia: two cases associated with IgG3 kappa cryoglobulin.
Am J Kidney Dis
2002
, vol. 
40
 
5
(pg. 
1091
-
1096
)
84
Payet
 
J
Livartowski
 
J
Kavian
 
N
, et al. 
Type I cryoglobulinemia in multiple myeloma, a rare entity: analysis of clinical and biological characteristics of seven cases and review of the literature.
Leuk Lymphoma
2013
, vol. 
54
 
4
(pg. 
767
-
777
)
85
Gorevic
 
PD
Kassab
 
HJ
Levo
 
Y
Kohn
 
R
Meltzer
 
M
Prose
 
P
Franklin
 
EC
Mixed cryoglobulinemia: clinical aspects and long-term follow-up of 40 patients.
Am J Med
1980
, vol. 
69
 
2
(pg. 
287
-
308
)
86
Ferri
 
C
Greco
 
F
Longombardo
 
G
, et al. 
Association between hepatitis C virus and mixed cryoglobulinemia [see comment]. [see comment]
Clin Exp Rheumatol
1991
, vol. 
9
 
6
(pg. 
621
-
624
)
87
Saadoun
 
D
Sellam
 
J
Ghillani-Dalbin
 
P
Crecel
 
R
Piette
 
JC
Cacoub
 
P
Increased risks of lymphoma and death among patients with non-hepatitis C virus-related mixed cryoglobulinemia.
Arch Intern Med
2006
, vol. 
166
 
19
(pg. 
2101
-
2108
)
88
Ramos-Casals
 
M
Stone
 
JH
Cid
 
MC
Bosch
 
X
The cryoglobulinaemias.
Lancet
2012
, vol. 
379
 
9813
(pg. 
348
-
360
)
89
Saadoun
 
D
Resche Rigon
 
M
Sene
 
D
, et al. 
Rituximab plus Peg-interferon-alpha/ribavirin compared with Peg-interferon-alpha/ribavirin in hepatitis C-related mixed cryoglobulinemia.
Blood
2010
, vol. 
116
 
3
(pg. 
326
-
334
)
90
Dammacco
 
F
Tucci
 
FA
Lauletta
 
G
, et al. 
Pegylated interferon-alpha, ribavirin, and rituximab combined therapy of hepatitis C virus-related mixed cryoglobulinemia: a long-term study.
Blood
2010
, vol. 
116
 
3
(pg. 
343
-
353
)
91
Terrier
 
B
Krastinova
 
E
Marie
 
I
, et al. 
Management of noninfectious mixed cryoglobulinemia vasculitis: data from 242 cases included in the CryoVas survey.
Blood
2012
, vol. 
119
 
25
(pg. 
5996
-
6004
)
92
Nasr
 
SH
Fidler
 
ME
Cornell
 
LD
, et al. 
Immunotactoid glomerulopathy: clinicopathologic and proteomic study.
Nephrol Dial Transplant
2012
, vol. 
27
 
11
(pg. 
4137
-
4146
)
93
Bridoux
 
F
Binaut
 
R
G.
 
Z
, et al. 
Glomerulopathy with non-organized and non-Randall type monoclonal immunoglobulin deposits A rare entity [Abstract].
J Am Soc Nephrol
2001
, vol. 
12
 
9
pg. 
94a
  
Abstract 492
94
Nasr
 
SH
Markowitz
 
GS
Stokes
 
MB
, et al. 
Proliferative glomerulonephritis with monoclonal IgG deposits: a distinct entity mimicking immune-complex glomerulonephritis.
Kidney Int
2004
, vol. 
65
 
1
(pg. 
85
-
96
)
95
Nasr
 
SH
Satoskar
 
A
Markowitz
 
GS
, et al. 
Proliferative glomerulonephritis with monoclonal IgG deposits.
J Am Soc Nephrol
2009
, vol. 
20
 
9
(pg. 
2055
-
2064
)
96
Guiard
 
E
Karras
 
A
Plaisier
 
E
, et al. 
Patterns of noncryoglobulinemic glomerulonephritis with monoclonal Ig deposits: correlation with IgG subclass and response to rituximab.
Clin J Am Soc Nephrol
2011
, vol. 
6
 
7
(pg. 
1609
-
1616
)
97
Lorenz
 
EC
Sethi
 
S
Leung
 
N
Dispenzieri
 
A
Fervenza
 
FC
Cosio
 
FG
Recurrent membranoproliferative glomerulonephritis after kidney transplantation.
Kidney Int
2010
, vol. 
77
 
8
(pg. 
721
-
728
)
98
Ma
 
CX
Lacy
 
MQ
Rompala
 
JF
, et al. 
Acquired Fanconi syndrome is an indolent disorder in the absence of overt multiple myeloma.
Blood
2004
, vol. 
104
 
1
(pg. 
40
-
42
)
99
Déret
 
S
Denoroy
 
L
Lamarine
 
M
, et al. 
Kappa light chain-associated Fanconi’s syndrome: molecular analysis of monoclonal immunoglobulin light chains from patients with and without intracellular crystals.
Protein Eng
1999
, vol. 
12
 
4
(pg. 
363
-
369
)
100
Bridoux
 
F
Sirac
 
C
Hugue
 
V
, et al. 
Fanconi's syndrome induced by a monoclonal Vkappa3 light chain in Waldenstrom's macroglobulinemia.
Am J Kidney Dis
2005
, vol. 
45
 
4
(pg. 
749
-
757
)
101
Lebeau
 
A
Zeindl-Eberhart
 
E
Müller
 
EC
Müller-Höcker
 
J
Jungblut
 
PR
Emmerich
 
B
Löhrs
 
U
Generalized crystal-storing histiocytosis associated with monoclonal gammopathy: molecular analysis of a disorder with rapid clinical course and review of the literature.
Blood
2002
, vol. 
100
 
5
(pg. 
1817
-
1827
)
102
El Hamel
 
C
Thierry
 
A
Trouillas
 
P
, et al. 
Crystal-storing histiocytosis with renal Fanconi syndrome: pathological and molecular characteristics compared with classical myeloma-associated Fanconi syndrome.
Nephrol Dial Transplant
2010
, vol. 
25
 
9
(pg. 
2982
-
2990
)
103
Dispenzieri
 
A
How I treat POEMS syndrome.
Blood
2012
, vol. 
119
 
24
(pg. 
5650
-
5658
)
104
Bridoux
 
F
Desport
 
E
Frémeaux-Bacchi
 
V
, et al. 
Glomerulonephritis with isolated C3 deposits and monoclonal gammopathy: a fortuitous association?
Clin J Am Soc Nephrol
2011
, vol. 
6
 
9
(pg. 
2165
-
2174
)
105
Sethi
 
S
Sukov
 
WR
Zhang
 
Y
, et al. 
Dense deposit disease associated with monoclonal gammopathy of undetermined significance.
Am J Kidney Dis
2010
, vol. 
56
 
5
(pg. 
977
-
982
)
Sign in via your Institution