Journal Information
Vol. 37. Issue. 5.September - October 2017
Pages 461-562
Visits
14113
Vol. 37. Issue. 5.September - October 2017
Pages 461-562
Review
Open Access
Monoclonal gammopathies of renal significance
Gammapatías monoclonales de significado renal
Visits
14113
Fernando Caravaca-Fontána,
Corresponding author
fcaravacaf@gmail.com

Corresponding author.
, Eduardo Gutiérreza, Ramón Delgado Lillob, Manuel Pragaa
a Servicio de Nefrología, Hospital Universitario 12 de Octubre, Madrid, Spain
b Servicio de Nefrología, Hospital Ruber Juan Bravo, Madrid, Spain
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Tables (6)
Table 1. Histological patterns of glomerular damage.
Table 2. Histological patterns of tubular damage.
Table 3. Histological patterns of mixed glomerular and tubular damage.
Table 4. Classification scheme of MGRS-associated disease according to the presence of organised or non-organised deposits.
Table 5. Techniques for the haematological and renal histology diagnosis of MGRSs.
Table 6. Therapeutic regimens proposed for MGRSs.
Show moreShow less
Abstract

The term monoclonal gammopathy of renal significance (MGRS) comprises a group of diseases pathogenetically characterised by proliferation of a B-cell or plasma cell clone that synthesises and secretes a monoclonal immunoglobulin or its components (light and/or heavy chains), that may deposit and cause glomerular, tubular, interstitial and/or vascular damage. The importance of differentiating the term MGRS from other monoclonal gammopathies lies in the fact that diagnostic and therapeutic procedures aimed at controlling monoclonal protein synthesis and secretion can be indicated, irrespective of the classic criteria based on malignant tumour expansion. Renal pathology associated with MGRS is highly heterogeneous, and therefore renal biopsy should be considered a key diagnostic tool. A precise diagnostic approach, however, must also identify the monoclonal protein in plasma and/or in urine, together with a complete haematological study in order to determine the nature and extension of cell clones. Recent advances in the understanding of these entities have resulted in significant improvements in clinical course and survival in several forms of MGRS, although more studies and clinical experience are needed in order to delineate more effective therapeutic strategies. In this review, we summarise the main clinical and pathological features of MGRS, highlighting the most appropriate diagnostic approach and current therapeutic options.

Keywords:
Chronic kidney disease
Glomerulonephritis
Monoclonal gammopathy of renal significance
Monoclonal protein
Resumen

Bajo el término gammapatías monoclonales de significado renal (GMSR) se engloban un conjunto de enfermedades que se caracterizan patogénicamente por la proliferación de un clon de linfocitos B o células plasmáticas que sintetizan y segregan una inmunoglobulina monoclonal o uno de sus componentes (cadenas ligeras o pesadas), con capacidad para depositarse y producir daño a nivel glomerular, tubular, intersticial o vascular. La importancia de discriminar el término GMSR radica en poder indicar procedimientos diagnósticos y terapéuticos dirigidos al control de la síntesis y secreción de las proteínas monoclonales independientemente de los criterios clásicos vinculados con la expansión tumoral maligna. La patología renal asociada a las GMSR es muy heterogénea, lo que confiere a la biopsia renal una consideración de prueba diagnóstica clave. La correcta investigación diagnóstica de una GMSR debe incluir, además, la identificación en plasma u orina de la proteína monoclonal y un estudio hematológico completo que determine la naturaleza y extensión del clon celular. Los avances en el conocimiento de estas entidades han permitido mejorar el curso evolutivo y la supervivencia en varias formas de GMSR, aunque son necesarios más estudios y experiencia clínica para delinear protocolos terapéuticos más efectivos. En la presente revisión se resumen las principales características clínico-patológicas de las GMSR, se detalla la aproximación diagnóstica más adecuada, así como las opciones terapéuticas disponibles en el momento actual.

Palabras clave:
Enfermedad renal crónica
Gammapatía monoclonal de significado renal
Glomerulonefritis
Proteína monoclonal
Full Text
Introduction

Several clinical entities are grouped under the name monoclonal gammopathies (MGs). They share a clonal proliferation of B-cells or plasma cells with the capacity to produce and secrete large amounts of a unique type of immunoglobulin or a constituent part of it (monoclonal component). The monoclonal component may be constituted of a heavy chain (normally γ chain and, less frequently, α, μ, δ, ¿ chains) along with a light chain (κ or λ), isolated light chains and, in exceptional circumstances, only heavy chains.1

The range of diseases, clinical manifestations and adverse health effects and persistence of entities is not only related to neoplastic cell proliferation, but also to the damage produced by the deposit of these monoclonal proteins in different organs, or through more complex pathogenic mechanisms, including autoimmunity, inflammation and fibrogenesis.2–4

In 2003, the International Myeloma Working Group2 revised the criteria for the diagnosis and classification of the clinical entities that are grouped under the term MG. In accordance with these criteria, there are four different entities:

  • 1.

    MG of undetermined significance (MGUS): monoclonal component <30g/l, with proliferation of plasma cells in bone marrow (<10%) and lack of clinical evidence of myeloma, lymphoma or amyloidosis.

  • 2.

    Asymptomatic or quiescent myeloma: monoclonal component ≥30g/l, with proliferation of bone marrow plasma cells ≥10%, but without evidence of organs or tissues involvement and, absence of the typical tetrad of hypercalcaemia, renal involvement, anaemia and bone lesions.

  • 3.

    Symptomatic myeloma which requires involvement of organs or tissues and which can also present as non-secretory (with no secretory component of monoclonal proteins). In 2014, the following additional criteria were incorporated: the presence of ≥60% plasma cells in bone marrow, a involved/uninvolved serum free light chain ratio ≥100, or the existence of more than one focal lesion using advanced imaging techniques (computed tomography [CT], magnetic resonance imaging [MRI] or positron emission tomography with 18F-fluorodeoxyglucose [PET-CT]).5

  • 4.

    Solitary bone plasmacytoma, extramedullary plasmacytoma and multiple solitary plasmacytomas.

Approximately 60% of all MGs are MGUS.6 In MGUS, a clone of B-cells or plasma cells, which are generally not neoplastic, produces and secretes small quantities of a monoclonal immunoglobulin or its components (light or heavy chains).7,8

This entity is a relatively common finding in the adult population (prevalence of 0.7% in the general population, increasing to 3% in those over the age of 50 and to 5% in those above the age of 70),8 with an annual standardised incidence of 4–15 cases per 100,000 according to different studies,9,10 but which may peak at 169 cases per 100,000 in individuals older than 80.10

It is estimated that in MGUSs there is neoplastic transformation (myeloma or lymphoma) in 1% annually.11–13 The factors which have proven to be determinant risk factors for neoplastic transformation are the following11–13: abnormal kappa (κ) to lambda (λ) free light chains ratio, different monoclonal immunoglobulin component (light or heavy chains) or of IgA type, and monoclonal protein concentration ≥15g/l. If these three factors are present, the risk of neoplastic progression reaches 58% in 20 years, while it is only 5% if none of these characteristics is present.13

In addition to the risk of neoplastic transformation, it has been demonstrated that these patients are also three to five times more likely to suffer from kidney diseases,14 and it has been observed in some studies that 23% of patients with light-chain MGUS have kidney disease.15

In the 1980s, there were already reports that the pathological scope of an MGUS was not only limited to neoplastic transformation, but that the synthesis and secretion of monoclonal (M) proteins could also cause other pathological processes with systemic effects which may develop by different pathogenic mechanisms.16–18 The kidney is one of the most commonly involved organs in the clinical course of an MGUS.

Renal involvement is very common in symptomatic myeloma, while the main pathogenic mechanism of myeloma-associated nephropathy is intratubular precipitation of monoclonal proteins produced by neoplastic cells (“cylinder nephropathy”).19–22 In this case, the secretion of large quantities of the M-protein are required to produce a massive precipitation, and the pathogenic key in this nephropathy is conditioned by the high burden and aggressiveness of the tumour.21,22

Descriptions of different pathological renal processes related to MGs have been increasingly reported. This led to the term MG of renal significance (MGRS)1,23–28 being adopted in order to distinguish and remove the uncertainty that exists on the benign progression of other MGs.

The importance of differentiating the term MGRS lies mainly in indicating targeted diagnostic and therapeutic procedures to control the synthesis and secretion of M-proteins — if it is confirmed that these are pathogenically linked to the nephropathy — irrespective of the classic haematological criteria which are more closely linked to the spread of the malignant tumour.3,4,22,26

In this review, the main clinical–pathological characteristics of MGRSs are described, the most appropriate diagnostic approach is detailed and the therapeutic advances and future perspectives are also discussed.

Kidney disease associated with monoclonal gammopathies

Kidney disease associated with MGRSs is highly heterogeneous, which means that the renal biopsy is considered a key diagnostic test.1,22,25–28 However, the concomitant presence of kidney disease of another aetiology may make the correct histological interpretation difficult in some cases and be a confounding factor.25

For a correct investigation and interpretation of the findings, the optical microscopy must be accompanied of immunofluorescence with a panel of antibodies against light chains and immunoglobulin isotypes, electron microscopy (EM), must also be performed as well.4,22,25,29 In some cases, more sensitive but complex techniques should also be used, such as immunoelectron microscopy (IEM),30 or laser microdissection followed by proteomic mass spectrometry,31–34 to confirm the composition of the deposits and where they are located.

Immunofluorescence is crucial in the diagnosis to establish the pathogenic link to blood dyscrasia and, therefore, the use of anti- (κ and λ) light-chain antibodies should be compulsory practice in the histological study of any renal biopsy.25

In cases in which there is no usable kidney tissue in the frozen sample for immunofluorescence, it is possible to carry out “rescue” techniques of the sample in paraffin (Pronase treatment) with a high likelihood of success for the immunohistochemical detection of light chains.35

Different methods to classify the MGRSs have been proposed.1,4 One method considers the predominant location of the kidney damage (glomerular, tubular or mixed),1 although in practice it is not uncommon for more than one entity to overlap in the same biopsy.36–39 In Tables 1–3, the main clinical entities and their histological findings are summarised according to the extent of the damage.

Table 1.

Histological patterns of glomerular damage.

Glomerular disease  Clinical manifestations  Optical microscopy  Immunofluorescence  Electron microscopy 
Proliferative glomerulonephritis with monoclonal Ig deposits  Proteinuria, variable microhaematuria, hypertension. Kidney disease.
Frequent C3 hypocomplementaemia 
MPGN.
Less common: mesangial proliferative, crescentic, sclerosing or diffuse proliferative 
IgG (G3>G1>G2) with restriction of κ or λ light chains in mesangium and capillary wall.
Less common: IgM or IgA.
C3 or C1q deposits. 
Double image of glomerular capillaries’ profile.
Mesangial and subendothelial electron-dense deposits. Less common: subepithelial or intramembranous 
Type 1 cryoglobulinaemia-associated glomerulonephritis  Arthralgia, arthritis, purpura, neuropathy.
Proteinuria, microhaematuria, kidney disease. Frequent hypertension. Frequent C3 and C4 hypocomplementaemia 
MPGN or endocapillary proliferative glomerulonephritis.
PAS+ intraluminal deposits 
Granular deposits in mesangium and capillaries.
Monoclonal IgG, IgM or IgA deposits (more common with κ chains). C3, C4 or C1q deposits. 
Subendothelial and intracapillary deposits. Frequently organised in fibrils, microtubules or “in fingerprint”. 
Fibrillary glomerulonephritis  Nephrotic proteinuria, microhaematuria and kidney disease. Rarely a rapidly progressive course  Mesangial proliferation or MPGN. Sometimes presence of crescents. Congo red negative  IgG (G4 and G1) most commonly polyclonal  10–30nm fibrils with random orientation in mesangium and capillaries 
Immunotactoid glomerulopathy  Nephrotic proteinuria, microhaematuria and kidney disease. Often associated with chronic lymphocytic leukaemia or lymphocytic lymphoma  MPGN or membranous glomerulonephritis. Less common: proliferative endocapillary  IgG (most common IgG1) with restriction of κ or λ light chains. C3 deposits.  30–90nm microtubules with subendothelial or subepithelial deposits 
C3 glomerulopathy/atypical haemolytic uraemic syndrome associated with monoclonal gammopathy  By indirect deregulation of the complement system's alternative pathway.
Proteinuria, nephrotic syndrome, microhaematuria, kidney disease, thrombotic microangiopathy 
MPGN, proliferative endocapillary, mesangial or crescentic glomerulonephritis.
Arterial thrombosis or glomerular capillaries 
Intense deposits of C3 in mesangium and capillaries. Absence or lack of other reactants.
Pronase treatment may be required to detect monoclonal Ig. 
Mesangial, intramembranous and subendothelial deposits in C3GN, and mesangial and intramembranous in DDD 

C3GN: C3 glomerulonephritis; DDD: dense deposit disease; Ig: immunoglobulin; MPGN: membranoproliferative glomerulonephritis; PAS: periodic acid–Schiff.

Adapted from: Sethi et al.1 and Bridoux et al.4

Table 2.

Histological patterns of tubular damage.

Tubular disease  Clinical manifestations  Optical microscopy  Immunofluorescence  Electron microscopy 
Cylinder nephropathy  Associated with MM in 90% of cases.
Acute deterioration of kidney function or progressive course. Presence of light chains in blood or urine 
Presence of eosinophilic cylinders in H&E stain, negative in PAS, with intense surrounding inflammatory reaction (neutrophils, lymphocytes).
More common in distal tubule 
Intense staining for a light chain (κ or λIntratubular electron-dense material 
Light chain proximal tubulopathy  Proteinuria and deterioration of kidney function. Fanconi syndrome sometimes associated with glycosuria, aminoaciduria and phosphaturia  Cytoplasmic inclusions in proximal tubular epithelium (crystalline or non-crystalline). Normal glomeruli. May be associated with tubular atrophy and interstitial fibrosis  Restriction for κ light chain in crystalline forms. Pronase treatment is sometimes required to detect light chains  Rectangular or rhomboidal crystals in lysosomes or free in the cytoplasm. In the non-crystalline forms, increased lysosomes with mottled appearance 
Crystal-storing histiocytosis  Non-nephrotic proteinuria or complete nephrotic syndrome. Kidney disease. There may also be medullary, pulmonary or corneal deposits. Associated most frequently with MM and lymphoproliferative processes  Predominantly interstitial infiltration of histiocytes with eosinophilic inclusions. Sometimes also in tubular epithelium and podocytes  Monoclonal Ig with restriction of κ light chain.
Pronase treatment is sometimes required to detect light chains 
Interstitial histiocytes filled with crystals. Less common in tubular epithelium and podocytes 

H&E: haematoxylin and eosin; Ig: immunoglobulin; MM: multiple myeloma; MPGN: membranoproliferative glomerulonephritis; PAS: periodic acid–Schiff.

Adapted from: Sethi et al.1 and Bridoux et al.4

Table 3.

Histological patterns of mixed glomerular and tubular damage.

Glomerular and tubular disease  Clinical manifestations  Optical microscopy  Immunofluorescence  Electron microscopy 
Ig-related amyloidosis (AL, AH, AHL)  Proteinuria and nephrotic syndrome, with varying degrees of renal failure. Rare microhaematuria  PAS and silver-negative deposits in glomeruli, vessels and interstitium. Congo red positive  AL more common with λ or κ light chains.
Heavy chains (γ), or heavy and light chains less common 
10–30nm fibrils with random orientation in glomeruli, vessels and interstitium 
Monoclonal Ig deposition disease (disease from deposits of light or heavy chains or a mixture of both)  Proteinuria, nephrotic syndrome and kidney disease. Presence of light chains and albumin in urine. Variable extrarenal manifestations due to Ig deposits in other organs  MPGN pattern, mesangial proliferative or nodular sclerosis. Thickened glomerular and tubular basement membranes. Congo red negative  Diffuse linear staining for light chain, heavy chain or both in glomerular and tubular basement membrane. Most common light chain: κ; Most common heavy chain: γ  Granular deposits in mesangium and glomerular and tubular basement membranes 

AH: heavy-chain amyloidosis; AHL: heavy- and light-chain amyloidosis; AL: light-chain amyloidosis; Ig: immunoglobulin; MPGN: membranoproliferative glomerulonephritis; PAS: periodic acid–Schiff.

Adapted from: Sethi et al.1 and Bridoux et al.4

The most accepted classification of lesions associated with MGRS is based on the distinction of the structure of deposits or inclusions according to whether these show an “organised” or “non-organised” configuration4,25 (Table 4).

Table 4.

Classification scheme of MGRS-associated disease according to the presence of organised or non-organised deposits.

Organised depositsFibrilsIg-related amyloidosis (AL, AH, AHL) 
Fibrillary glomerulonephritis 
MicrotubulesImmunotactoid glomerulonephritis 
Type 1 cryoglobulinaemic glomerulonephritis 
Crystals or inclusionsProximal tubulopathy (with or without Fanconi syndrome) 
Crystal-storing histiocytosis 
Non-organised depositsMonoclonal Ig deposition disease (light or heavy chains or a mixture of both)
Proliferative glomerulonephritis with monoclonal Ig deposits
Monoclonal Ig-associated C3 glomerulonephritis

AH: heavy-chain amyloidosis; AHL: heavy- and light-chain amyloidosis; AL: light-chain amyloidosis; Ig: immunoglobulin.

Source: A modified version of the original scheme by Bridoux et al.4

“Organised” deposits are subdivided into: fibrils, microtubules and crystals or inclusions.

The pathological processes associated with fibrils are: immunoglobulin-related amyloidosis (of light chains, heavy chains or mixtures of light and heavy chains)4,25,40–43 and fibrillary glomerulonephritis.4,33,44,45

When the microstructure of the deposits adopts a microtubular structure, two entities can be distinguished4,25: immunotactoid glomerulonephritis, also known as glomerulonephritis with organised microtubules and monoclonal immunoglobulin deposits,33,44,46 and type 1 cryoglobulinaemia-associated glomerulonephritis.33,47–51

The crystal and inclusion deposits4,25 produce a predominantly tubular and interstitial disease, and two subtypes can be distinguished: proximal tubulopathy (with or without Fanconi syndrome)52–54 and histiocytosis with crystals stores,51,55–59 in which the crystal deposits are not found in the tubular epithelial cells, but instead inside the histiocytes.1 Cases of acute interstitial nephritis unrelated to the crystal deposits or intratubular precipitation of cylinders have also been reported, but with light chain deposits shown in the tubular basement membranes.60

Disease entities with “non-organised” deposits include: Randall-type monoclonal immunoglobulin deposit disease (disease caused by deposits of light chains, heavy chains or a combination of both)25,61–63; proliferative glomerulonephritis with monoclonal immunoglobulin deposits17,25,64–68 and monoclonal gammapathy-associated C3 glomerulopathy (C3G).69–74 In the case of proliferative glomerulonephritis with monoclonal immunoglobulin deposits, involvement is limited to the glomerulus, while in monoclonal immunoglobulin deposit disease there is extraglomerular and, frequently, extrarenal involvement.4

In addition to these processes, there are reports suggesting a possible pathogenic involvement of MG in other glomerulopathies, such as membranous GN,75,76 focal segmental glomerulosclerosis,77 pauci-immune extracapillary glomerulonephritis,78 C4 proliferative glomerulonephritis79 and in thrombotic microangiopathies.79–83

Clinical presentation

MGRSs may show a wide range of manifestations, depending on the underlying pathogenic mechanism and the primary site of involvement.1,3,4 In the majority of cases, the deposit of M-proteins is responsible for the kidney disease, while in other cases it is caused indirectly through a deregulation of the complement system's alternative pathway, resulting in C3G,1,3,4,71 or, more rarely, in atypical haemolytic uraemic syndrome.81 Thus, the monoclonal component would be able to interfere with the regulatory proteins of the complement system, acting as miniautoantibodies against factor H,73,74 or as a C3 nephritic factor, which stabilises the C3 convertase of the alternative pathway and maintains its hyperactivation.1,70,71

The structural and chemical characteristics of each specifc M-protein, and the inflammatory response of each individual, seem to be key to determine the type of kidney damage being produced.18,84 High-molecular-weight proteins, such as immunoglobulins (formed by heavy and light chains), do not cross the filtration barrier and are deposited in the glomerulus which triggers an inflammatory processes. In contrast, light chains are able to cross the filtration barrier and generate a variety of tubular damage.

Renal involvement is often the first manifestation of blood dyscrasia.26 According to the different series, the age of diagnosis tends to be over 50. The disease of monoclonal immunoglobulin deposits is more common in males and proliferative glomerulonephritis with monoclonal immunoglobulin deposits is more common in women.63,66,85

Within the clinical manifestations of MGRSs, it is common to find different degrees of proteinuria, which may reach the nephrotic range, together with microhaematuria and hypertension in certain cases. Renal failure is detected in a high percentage of patients at the time of diagnosis, which may progress to end-stage kidney disease.4,63,66,86 This is of particular importance given that the recurrence of some of these diseases in transplanted kidneys is very common.87–92 This fact illustrate the need for a correct diagnosis, even when it is unlikely to preserve the function of the native kidneys.

Extrarenal manifestations may also be presented, especially in cases of amyloidosis, monoclonal immunoglobulin deposition disease and type 1 cryoglobulinaemia, with involvement predominantly of the heart, liver, skin and joints.1,4,93 The possibility of the disease spreading to other organs and tissues in patients diagnosed with amyloidosis should be investigated,43 due to the common nature of cardiac involvement, which tends to be the main determinant of mortality.43

Cases of osteomalacia secondary to Fanconi syndrome have also been reported.52

Other systemic manifestations of an MG may be related to endothelial damage and systemic thrombotic microangiopathy associated with the secretion of vascular endothelial growth factor (VEGF), as occurs in POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, MG and skin lesions)94 and scleromyxoedema.95

Diagnosis of monoclonal gammopathies of renal significance

The appropriate diagnostic procedure of an MGRS should include, in addition to a renal biopsy, the demonstration and identification of MG in plasma or urine, a haematological study which determines the nature and extent of the cell clone causing the MG and, in some processes (e.g. amyloidosis), extension of the study to determine the spreading of the disease to other organs.4,25,26 The main techniques for the haematological and histological diagnosis are summarised in Table 5.

Table 5.

Techniques for the haematological and renal histology diagnosis of MGRSs.

Diagnosis  Haematological  Renal histology 
Recommended procedures  -Electrophoresis on plasma or urine
-Immunofixation on plasma or urine
-Free light chains in blood and urine
-Bone marrow aspiration/biopsy 
-Optical microscopy
-Immunofluorescence with staining of light chains (κ and λ) and Pronase treatment in selected cases
-Electron microscopy 
Additional diagnostic proceduresa  -Western blot technique for detection of IgG subclasses
-Imaging test: CT, MRI or PET-CT
-Lymph node biopsy 
-Immunoelectron microscopy
-Laser microdissection and mass spectrometry 

CT: computed tomography; IgG: immunoglobulin G; MRI: magnetic resonance imaging; PET-CT: positron emission tomography with 18F-fluorodeoxyglucose.

a

Depending on the diagnostic suspicion and clinical course, as well as availability in the centre.

The diagnosis of a suspected MGRS is almost always established by associating renal involvement (deterioration of function, proteinuria, Fanconi syndrome or other metabolic abnormalities associated with tubulointerstitial dysfunction) together with the presence of a monoclonal peak in the electrophoretic spectrum.4,96,97

In most cases, the diagnosis of MG is performed using conventional electrophoresis on plasma or urine.4,96 The presence of monoclonal immunoglobulins is usually identified by a tall narrow peak in the beta or gamma region, unlike polyclonal increase, which tends to create a wide band in the gamma region.96

However, in some cases the concentration of monoclonal protein in plasma or urine is so small that electrophoresis is unable to detect it. In fact, some cases of renal involvement in MG are diagnosed mainly by the findings in the renal biopsy (immunohistochemistry), without being suspected when the biopsy was indicated.

In addition to conventional electrophoresis, plasma and urine immunofixation must be performed in all cases to identify the type of M-protein, as this is more sensitive than electrophoresis in detecting the M-protein.4,96,98

Another diagnostic method is the determination of free light chains (FLCs) in blood and urine.96,98–102 The concentration of these proteins can be measured by a nephelometric immunoassay using polyclonal antibodies targeting light chain epitopes, which are exposed when the chain is in its free form but hidden when the chain is bound, creating the structure of the Ig.99

These measurements of FLCs are very sensitive, but the drawback is that they are not able to demonstrate the monoclonality of FLCs. This possibility is suggested indirectly by the ratio between the concentrations of the κ and λ chains.98,100

An abnormal ratio of κ and λ concentrations should be interpreted more carefully in patients with deterioration of kidney function.4,102 There is a strong correlation between the FLC concentration and kidney function; as the glomerular filtration rate decreases, the polyclonal FLC concentrations, of both κ and λ increase. Furthermore, in patients with normal kidney function, the greater physiological production of polyclonal κ FLC is masked by the more rapid clearance of monomeric forms of κ FLC, compared to the larger dimeric λ forms. This is how a change is produced in the ratio of κ and λ FLC concentrations in the case of renal failure.4,102 When kidney function is normal, the range of this ratio ranges from 0.26 to 1.65, while when kidney failure is present, the ratio accepted as normal ranges between 0.37 and 3.17, however a range for each stage of kidney failure has not been established.102

The difference in concentrations between FLCs is very useful not only for diagnosis, but also for follow-up and as a measure of the response to treatment. It is therefore recommended that it is monitored frequently.103

FLCs are also currently included in the treatment response criteria for AL amyloidosis.104 Therefore, a normalisation of the ratio of κ and λ concentrations, together with a negative result in the blood and urine immunofixation, is needed to be certain of a complete remission.

Using the FLCs measurement as an isolated test for detecting an MG is subject to debate105 and, although it may be useful to support the diagnosis of MG in myeloma, macroglobulinaemia and amyloidosis, the current recommendations state that electrophoresis (EP) and blood and urine immunofixation should be used for the diagnosis of MG.

More complex tests (western blot or electrotransfer) on blood and urine are required for the detection of some subclasses of immunoglobulins (IgG) or circulating monoclonal incomplete heavy chains. These tests are able to detect very small concentrations of monoclonal proteins with a high sensitivity,106 unfortunately these techniques are not routinely available.

In order to confirm the diagnosis of MGRS, it is not only necessary to prove the pathogenic involvement of MG in the kidney, but, in addition, myeloma must be ruled out and the cell clone producing the M-protein must be characterised because its relevance in the design of the therapeutic strategy. The support of a Haematology Department is needed for this.3,4,107

In cases of MGRS in which the renal biopsy shows IgG, IgA or LC, the bone marrow aspiration and biopsy use to be sufficient to demonstrate the clone using flow cytometry and immunohistochemical techniques.4 In addition, the spread of the bone tumour or the presence of solitary plasmacytoma should be ruled out with imaging techniques (CT, MRI or PET-CT).4,5 MRI is the technique with the greatest sensitivity in the detection of bone marrow infiltration, although it is laborious.5 PET-CT, on the other hand, is able to identify changes in the lesions throughout the follow up and avoids the use of intravenous contrast. However, it exposes patients to high levels of radiation and is more expensive.5

The possibility that a clone is from B-cells instead of from plasma cells should be suspected in patients with IgM MGRS. Then, the imaging study include areas suspected of hosting lymphadenopathies for biopsy.3

Treatment

The treatment strategies for MGRSs are based on chemotherapy that must be adapted to the nature of the cell clone, both for lymphocytes and plasma cells, to the kidney function and to the presence or non-presence of extrarenal involvement.22,107

The rapid suppression of nephrotoxic monoclonal immunoglobulin has proven to be a treatment with satisfactory results on kidney function and patient survival in several forms of MGRS.107 However, as will be emphasised later, more studies and clinical experience are required to obtain therapeutic protocols based on solid evidence.

The fundamental objective of treatment, except for the case of light chain amyloidosis, must be aimed at preserving kidney function.26,91,107,108 This means that patients with irreversible kidney damage would not be candidates to receive chemotherapy treatment, unless there is a desire to obtain complete haematological remission to prevent recurrence in a kidney transplant.107

In 2013, the International Kidney and Monoclonal Gammopathy Working Group published a consensus document on the treatment regimens recommended for MGRSs, based on the clinical experience of the treatment of malignant blood dyscrasias.107Table 6 summarises the group's main recommendations. However, given the heterogeneity of MGRS-associated diseases, there is uncertainty regarding the optimal treatment for some more complex presentations with a clinical experience that is limited to isolated case reports or small series of cases.63,64,70,71,77,109 Thus, different regimens have been used for fibrillary glomerulonephritis, including cyclophosphamide, mycophenolate mofetil, cyclosporine, melphalan, lenalidomide and rituximab, with limited results.44,45,110 Furthermore, treatment regimens similar to those used for proliferative glomerulonephritis with monoclonal immunoglobulin deposits have been used for monoclonal gammopathy-associated C3 glomerulopathy.70,71,107 However, the information available on the long-term prognosis of these processes using the current therapies is insufficient.26

Table 6.

Therapeutic regimens proposed for MGRSs.

Disease  Treatment 
Proliferative glomerulonephritis with monoclonal Ig depositsStage 1–2 CKD, proteinuria <1g/day: observation 
Stage 1–2 CKD with proteinuria >1g/day, progressive CKD or stage 3–4 CKD: 
-Cyclophosphamide+bortezomib+dexamethasone if IgG or IgA 
-Rituximab±cyclophosphamide+dexamethasone if IgM 
-If <65 years: high-dose melphalan following an HSCT 
Stage 5 CKD candidate for kidney transplant: high-dose melphalan+HSCT 
Type 1 cryoglobulinaemia-associated glomerulonephritisPaucisymptomatic or low-grade B-cell proliferation: observation 
Symptomatic: 
-Plasmacytic clone: bortezomib+cyclophosphamide±thalidomide 
-Lymphoplasmacytic clone: rituximab 
-Alternative: bendamustine 
Immunotactoid glomerulopathyTreatment regimens similar to those employed in CLL, based on cyclophosphamide or bendamustine±rituximab 
For isolated gammopathy, treatment regimen based on bortezomib 
Ig-related amyloidosis (AL, AH, AHL)Classification in three stages according to the increase in levels of NT-proBNP and troponin 
Stage 1–2: melphalan or cyclophosphamide+dexamethasone+bortezomib 
Stage 3: cyclophosphamide+dexamethasone+bortezomib 
Heart transplant if cardiac involvement; HSCT in selected cases 
Monoclonal Ig deposition diseaseStage 1–3 CKD: cyclophosphamide+bortezomib+dexamethasone, with subsequent HSCT in selected cases. Alternative: bendamustine 
Stages 4–5 CKD: cyclophosphamide+bortezomib+dexamethasone+HSCT if candidate for kidney transplant 
Light chain proximal tubulopathyStage 1–3 CKD: cyclophosphamide, bortezomib or thalidomide+HSCT in selected cases 
Stage 4–5 CKD: HSCT if they are candidates for a kidney transplant. If not, conservative management 

AH: heavy-chain amyloidosis; AHL: heavy- and light-chain amyloidosis; AL: light-chain amyloidosis; CKD: chronic kidney disease; HSCT: haematopoietic stem cell transplantation; Ig: immunoglobulin; NT-proBNP: N-terminal pro-brain natriuretic peptide.

Source: Adapted from original by Fermand et al.107

Due to the growing importance of MGRSs in clinical nephrology, it is essential to be aware of the main therapeutic agents which are currently used, their tolerance and the most common side effects. Many of these treatments are administered in a combined form due to the synergistic effect on the B-cells and plasma cells.111 The main combinations include107,111,112: bortezomib, cyclophosphamide and dexamethasone; bendamustine and rituximab, and immunomodulatory agents, such as thalidomide and lenalidomide.

Bortezomib has a pivotal role within the therapeutic arsenal because of its safety profile and the possibility of being administered in full doses to patients with advanced kidney disease.112–114 The mechanism of action is based on the inhibition of proteasome activity, which produces apoptosis of the plasma cell and also inhibits the NF-κB pathway by reducing the release of proinflammatory cytokines and inducing anti-apoptotic pathways at the tubular level.115–117 Usually the side effects are not serious; it causes development or worsening of peripheral neuropathy, although this is less common when the route of administration is subcutaneous.118 Prophylaxis against herpes zoster is also recommended due to the risk of reactivation.111

Among the cytotoxic agents, both melphalan and cyclophosphamide have an effect on B-cells and plasma cells, however cyclophosphamide is used more frequently because it is less toxic.107,111 Another alternative is bendamustine, approved for the treatment of some lymphomas and suitable for patients with renal failure since it is predominantly metabolised in the liver.119–121

Melphalan is used in myeloablative doses as a conditioning therapy for autologous haematopoietic stem cell transplantation in selected cases with no significant extrarenal disease.107,111 This autologous transplant has been proven to improve survival of patients with multiple myeloma and light chain amyloidosis. However, in practice less than 20% of patients are suitable candidates for this treatment, which is associated with high morbidity and mortality.86,122–124

Monoclonal antibodies such as rituximab (targeting the CD20 antigen) are a suitable therapeutic option in the different forms of B-cell mediated MGRS, because of their good tolerance and limited number of side effects.107,111,112 The use of daratumumab (targeting CD38) for relapsed or refractory myeloma has been approved.125 However, experience on its use in cases of MGRS is currently lacking.

Within the family of immunomodulatory drugs, thalidomide would be more suitable than lenalidomide, due to the fact that the latter is excreted by the kidneys and can also cause kidney function deterioration in some cases.126–128 However, side effects have also been reported with thalidomide, such as the development of hyperkalaemia.129

Conclusions

MGRSs are associated with a wide range of kidney diseases resulting from the depositions of immunoglobulin or of its components in the kidneys, or through the deregulation of the complement system. Although the mortality of patients with MGRS is lower than that of myeloma or other related neoplastic forms, the likelihood of developing advanced chronic kidney disease is very high. For this reason, when evaluating patients with suspected MGRS it is necessary to conduct complete anatomopathological, haematological and biochemical studies. These studies allow to determine the type of entity and the extension of the disese. Advances in the understanding of these entities have made it possible to improve the clinical course and survival in several forms of MGRS. However, more studies and clinical experience are necessary to design more effective therapeutic protocols. It is a priority the close collaboration between nephrologists and haematologists to individualise the treatment to the clinical characteristics and comorbidity of patients, in an attempt to improve the overall prognosis of these diseases.

The incidence and prevalence of this group of diseases in the Spanish population is not currently known. There are few cases published.130 According to the clinical experience, these diseases are not frequent, but so far no epidemiological studies have been conducted which confirm these assessments. Furthermore, as they are rare diseases, but requiring a complex study, the diagnosis might end up being less appropriate or incomplete in patients attended to in many hospitals which do not have the necessary non-conventional diagnostic tools. Nevertheless the implementation of these tools in all hospitals does not seem to be an efficient measure. Therefore, the creation of centres of excellence and diagnostic–therapeutic reference for these types of diseases could be an appropriate investment to benefit from the health care, academic and economic value.

GLOSEN study proposal

In 2009, an international group for research into MGRSs was created. Nephrology, haematology and anatomical pathology departments from several countries participate in the group.4,26,107 The studies and publications by the members of this research group are currently recognised as the forefront of research into these diseases, with very significant diagnostic and therapeutic advances. The incorporation of Spanish groups into this international group could result in mutual benefits, such as training and transfer of experience to Spanish members, as well as the incorporation of Spanish centres and patients into international clinical trials.

All of these reasons could justify the creation of a national MGRS register which would help to research the clinical–pathological characteristics of these entities, and to identify evolutionary and response determinants to current treatments.

We believe that the ‘Grupo de Estudio de la Patología Glomerular de la Sociedad Española de Nefrología’ [Glomerular Disease Study Group of the Spanish Society of Nephrology] (GLOSEN) may be the ideal framework to lead a study of these characteristics, given its ample experience in conducting collaborative projects.131–133

As an initial project, it is proposed to collect retrospectively all diagnosed cases of MGRS with renal biopsy during recent years from the different centres taking part in the study. Although a work proposal will be sent to all members of the group soon, we are hereby launching a message to attract the interest of all nephrologists and pathologists interested in glomerular diseases, and requesting their collaboration in the study.

Conflicts of interest

The authors declare that they have no conflicts of interest related to the publication of this article.

Key concepts

  • 1.

    MGRSs are characterised by the proliferation of a clone of B-cells or plasma cells which synthesise and secrete a monoclonal immunoglobulin or one of its components (light or heavy chains), with the capacity to be deposited and be the cause glomerular, tubular, interstitial or vascular damage.

  • 2.

    Given the heterogeneity of MGRS-associated kidney disease, the renal biopsy is fundamental, and its proper histological analysis must include optical microscopy, immunofluorescence and electron microscopy.

  • 3.

    There are different ways of classifying MGRS-associated kidney disease, although the most accepted method is in accordance with the organisation of the deposits: “organised” (fibrils, microtubules and crystals) or “non-organised” (monoclonal immunoglobulin deposition disease, proliferative glomerulonephritis with monoclonal immunoglobulin deposits and monoclonal immunoglobulin-associated C3 glomerulonephritis).

  • 4.

    The diagnostic study should also include electrophoresis and plasma and urine immunofixation to identify the monoclonal protein and the determination of free light chains.

  • 5.

    In addition, myeloma should be ruled out and the cell clone producing the monoclonal protein should be characterised by a bone marrow aspiration and biopsy.

  • 6.

    Current treatment is based on clinical experience of the treatment of malignant blood dyscrasias. Close collaboration between nephrologists and haematologists to personalise the treatment to the clinical characteristics and comorbidity of patients is therefore a priority.

Acknowledgements

This study has been partly supported by the FEDER [Spanish Federation for Rare Diseases] funds ISCIII-RETIC REDinREN RD16/0009.

References
[1]
S. Sethi, F.C. Fervenza, S.V. Rajkumar.
Spectrum of manifestations of monoclonal gammopathy-associated renal lesions.
Curr Opin Nephrol Hypertens, 25 (2016), pp. 127-137
[2]
International Myeloma Working Group.
Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report of the International Myeloma Working Group.
Br J Haematol, 121 (2003), pp. 749-757
[3]
S.V. Glavey, N. Leung.
Monoclonal gammopathy: the good, the bad and the ugly.
Blood Rev, 30 (2016), pp. 223-231
[4]
F. Bridoux, N. Leung, C.A. Hutchison, G. Touchard, S. Sethi, J.P. Fermand, et al.
International Kidney and Monoclonal Gammopathy Research Group: Diagnosis of monoclonal gammopathy of renal significance.
Kidney Int, 87 (2015), pp. 698-711
[5]
G. Pratt, S. Bowcock, A. Chantry, G. Cook, G. Jackson, M. Lai, et al.
Time to redefine myeloma.
Br J Haematol, 171 (2015), pp. 1-10
[6]
R.A. Kyle, S.V. Rajkumar.
Monoclonal gammopathies of undetermined significance.
Best Pract Res Clin Haematol, 18 (2005), pp. 689-707
[7]
R.A. Kyle.
Monoclonal gammopathy of undetermined significance (MGUS).
Baillieres Clin Haematol, 8 (1995), pp. 761-781
[8]
R.A. Kyle, T.M. Therneau, S.V. Rajkumar, D.R. Larson, M.F. Plevak, J.R. Offord, et al.
Prevalence of monoclonal gammopathy of undetermined significance.
N Engl J Med, 354 (2006), pp. 1362-1369
[9]
Q. Cabrera, M. Macro, B. Hebert, E. Cornet, A. Collignon, X. Troussard.
Epidemiology of monoclonal gammopathy of undetermined significance (MGUS): the experience from the specialized registry of hematologic malignancies of Basse-Normandie (France).
Cancer Epidemiol, 38 (2014), pp. 354-356
[10]
H.M. Ogmundsdóttir, V. Haraldsdóttir, M.G. Jóhannesson, G. Olafsdóttir, K. Bjarnadóttir, H. Sigvaldason.
Monoclonal gammopathy in Iceland: a population-based registry and follow-up.
Br J Haematol, 118 (2002), pp. 166-173
[11]
J. Bladè, L. Rosiñol, M.T. Cibeira, C.F. de Larrea.
Pathogenesis and progression of monoclonal gammopathy of undetermined significance.
Leukemia, 22 (2008), pp. 1651-1657
[12]
R.A. Kyle, B.G. Durie, S.V. Rajkumar, O. Landgren, J. Blade, G. Merlini, et al.
Monoclonal gammopathy of undetermined significance (MGUS) and smoldering (asymptomatic) multiple myeloma: IMWG consensus perspectives risk factors for progression and guidelines for monitoring and management.
Leukemia, 24 (2010), pp. 1121-1127
[13]
R.A. Kyle, S.V. Rajkumar.
Monoclonal gammopathy of undetermined significance and smouldering multiple myeloma: emphasis on risk factors for progression.
Br J Haematol, 139 (2007), pp. 730-743
[14]
S.Y. Kristinsson, M. Björkholm, T.M. Andersson, S. Eloranta, P.W. Dickman, L.R. Goldin, et al.
Patterns of survival and causes of death following a diagnosis of monoclonal gammopathy of undetermined significance: a population-based study.
Haematologica, 94 (2009), pp. 1714-1720
[15]
A. Dispenzieri, J.A. Katzmann, R.A. Kyle, D.R. Larson, L.J. Melton, C.L. Colby, et al.
Prevalence and risk of progression of light-chain monoclonal gammopathy of undetermined significance: a retrospective population-based cohort study.
Lancet, 375 (2010), pp. 1721-1728
[16]
C.E. Alpers, J. Hopper Jr., C.G. Biava.
Light-chain glomerulopathy with amyloid-like deposits.
Hum Pathol, 15 (1984), pp. 444-448
[17]
C.E. Alpers, W.H. Tu, J. Hopper Jr., C.G. Biava.
Single light chain subclass (kappa chain) immunoglobulin deposition in glomerulonephritis.
Hum Pathol, 16 (1985), pp. 294-304
[18]
A. Solomon, D.T. Weiss, A.A. Kattine.
Nephrotoxic potential of Bence Jones proteins.
N Engl J Med, 324 (1991), pp. 1845-1851
[19]
R. Alexanian, B. Barlogie, D. Dixon.
Renal failure in multiple myeloma pathogenesis and prognostic implications.
Arch Intern Med, 150 (1990), pp. 1693-1695
[20]
L.M. Knudsen, M. Hjorth, E. Hippe.
Renal failure in multiple myeloma: reversibility and impact on the prognosis Nordic Myeloma Study Group.
Eur J Haematol, 65 (2000), pp. 175-181
[21]
A. Davenport, G. Merlini.
Myeloma kidney: advances in molecular mechanisms of acute kidney injury open novel therapeutic opportunities.
Nephrol Dial Transplant, 27 (2012), pp. 3713-3718
[22]
E.C. Heher, H.G. Rennke, J.P. Laubach, P.G. Richardson.
Kidney disease and multiple myeloma.
Clin J Am Soc Nephrol, 8 (2013), pp. 2007-2017
[23]
P. Paueksakon, M.P. Revelo, R.G. Horn, S. Shappell, A.B. Fogo.
Monoclonal gammopathy: significance and possible causality in renal disease.
Am J Kidney Dis, 42 (2003), pp. 87-95
[24]
G. Merlini, M.J. Stone.
Dangerous small B-cell clones.
Blood, 108 (2006), pp. 2520-2530
[25]
G.A. Herrera.
Renal lesions associated with plasma cell dyscrasias: practical approach to diagnosis, new concepts, and challenges.
Arch Pathol Lab Med, 133 (2009), pp. 249-267
[26]
N. Leung, F. Bridoux, C.A. Hutchison, S.H. Nasr, P. Cockwell, J.P. Fermand, et al.
International Kidney and Monoclonal Gammopathy Research Group Monoclonal gammopathy of renal significance: when MGUS is no longer undetermined or insignificant.
Blood, 120 (2012), pp. 4292-4295
[27]
T. Al-Hussain, M.H. Hussein, H.A. Mana, M. Akhtar.
Renal involvement in monoclonal gammopathy.
Adv Anat Pathol, 22 (2015), pp. 121-134
[28]
S. Chauvet, F. Bridoux, L. Ecotière, V. Javaugue, C. Sirac, B. Arnulf, et al.
Kidney diseases associated with monoclonal immunoglobulin M-secreting B-cell lymphoproliferative disorders: a case series of 35 patients.
Am J Kidney Dis, 66 (2015), pp. 756-767
[29]
G.A. Herrera.
The contributions of electron microscopy to the understanding and diagnosis of plasma cell dyscrasia-related renal lesions.
Med Electron Microsc, 34 (2001), pp. 1-18
[30]
G.A. Herrera, E.A. Turbat-Herrera.
Ultrastructural immunolabeling in the diagnosis of monoclonal light-and heavy-chain-related renal diseases.
Ultrastruct Pathol, 34 (2010), pp. 161-173
[31]
S. Sethi, J.A. Vrana, J.D. Theis, N. Leung, A. Sethi, S.H. Nasr, et al.
Laser microdissection and mass spectrometry-based proteomics aids the diagnosis and typing of renal amyloidosis.
Kidney Int, 82 (2012), pp. 226-234
[32]
S.H. Nasr, S.M. Said, A.M. Valeri, S. Sethi, M.E. Fidler, L.D. Cornell, et al.
The diagnosis and characteristics of renal heavy-chain and heavy/light-chain amyloidosis and their comparison with renal light-chain amyloidosis.
Kidney Int, 83 (2013), pp. 463-470
[33]
S. Sethi, J.D. Theis, J.A. Vrana, F.C. Fervenza, A. Sethi, Q. Qian, et al.
Laser microdissection and proteomic analysis of amyloidosis, cryoglobulinemic GN, fibrillary GN, and immunotactoid glomerulopathy.
Clin J Am Soc Nephrol, 8 (2013), pp. 915-921
[34]
D. Jain, J.A. Green, S. Bastacky, J.D. Theis, S. Sethi.
Membranoproliferative glomerulonephritis: the role for laser microdissection and mass spectrometry.
Am J Kidney Dis, 63 (2014), pp. 324-328
[35]
S.H. Nasr, S.J. Galgano, G.S. Markowitz, M.B. Stokes, V.D. D’Agati.
Immunofluorescence on pronase-digested paraffin sections: a valuable salvage technique for renal biopsies.
Kidney Int, 70 (2006), pp. 2148-2151
[36]
N. Leung, S.H. Nasr.
A patient with abnormal kidney function and a monoclonal light chain in the urine.
Clin J Am Nephrol, 11 (2016), pp. 1073-1082
[37]
S.H. Nasr, A.M. Valeri, S. Sethi, M.E. Fidler, L.D. Cornell, M.A. Gertz, et al.
Clinicopathologic correlations in multiple myeloma: a case series of 190 patients with kidney biopsies.
Am J Kidney Dis, 59 (2012), pp. 786-794
[38]
E.C. Lorenz, S. Sethi, T.L. Poshusta, M. Ramirez-Alvarado, S. Kumar, D.J. Lager, et al.
Renal failure due to combined cast nephropathy, amyloidosis and light-chain deposition disease.
Nephrol Dial Transplant, 25 (2010), pp. 1340-1343
[39]
Q. Qian, N. Leung, J.D. Theis, A. Dogan, S. Sethi.
Coexistence of myeloma cast nephropathy, light chain deposition disease, and nonamyloid fibrils in a patient with multiple myeloma.
Am J Kidney Dis, 56 (2010), pp. 971-976
[40]
C. Pozzi, F. Locatelli.
Kidney and liver involvement in monoclonal light chain disorders.
Semin Nephrol, 22 (2002), pp. 319-330
[41]
M.M. Picken.
Amyloidosis-where are we now and where are we heading.
Arch Pathol Lab Med, 134 (2010), pp. 545-551
[42]
S.M. Said, S. Sethi, A.M. Valeri, N. Leung, L.D. Cornell, M.E. Fidler, et al.
Renal amyloidosis: origin and clinicopathologic correlations of 474 recent cases.
Clin J Am Soc Nephrol, 8 (2013), pp. 1515-1523
[43]
G. Merlini, A.D. Wechalekar, G. Palladini.
Systemic light chain amyloidosis: an update for treating physicians.
Blood, 121 (2013), pp. 5124-5130
[44]
J.L. Rosenstock, G.S. Markowitz, A.M. Valeri, G. Sacchi, G.B. Appel, V.D. D’Agati.
Fibrillary and immunotactoid glomerulonephritis: distinct entities with different clinical and pathologic features.
Kidney Int, 63 (2003), pp. 1450-1461
[45]
S.H. Nasr, A.M. Valeri, L.D. Cornell, M.E. Fidler, S. Sethi, Leung., et al.
Fibrillary glomerulonephritis: a report of 66 cases from a single institution.
Clin J Am Soc Nephrol, 6 (2011), pp. 775-784
[46]
S.H. Nasr, M.E. Fidler, L.D. Cornell, N. Leung, F.G. Cosio, S.S. Sheikh, et al.
Immunotactoid glomerulopathy: clinicopathologic and proteomic study.
Nephrol Dial Transplant, 27 (2012), pp. 4137-4146
[47]
B. Terrier, A. Karras, J.E. Kahn, G. Le Guenno, I. Marie, L. Benarous, et al.
The spectrum of type I cryoglobulinemia vasculitis: new insights based on 64 cases.
Medicine (Baltimore), 92 (2013), pp. 61-68
[48]
S.H. Nasr, G.S. Markowitz, B.S. Reddy, J. Maesaka, M.A. Swidler, V.D. D’Agati.
Dysproteinemia, proteinuria, and glomerulonephritis.
Kidney Int, 69 (2006), pp. 772-775
[49]
A. Karras, L.H. Noël, D. Droz, D. Delansorne, J.P. Saint-André, P. Aucouturier, et al.
Renal involvement in monoclonal (type I) cryoglobulinemia: two cases associated with IgG3 kappa cryoglobulin.
Am J Kidney Dis, 40 (2002), pp. 1091-1096
[50]
J. Hemminger, M. Kandarpa, A. Tsai, T. Nadasdy.
Proliferative glomerulonephritis with monoclonal IgG1 deposits in a hepatitis C virus-positive patient.
Am J Kidney Dis, 67 (2016), pp. 703-708
[51]
P.A. DeLyria, S.E. Avedschmidt, C. Yamada, E.A. Farkash.
Fatal cryocrystalglobulinemia with intravascular and renal tubular crystalline deposits.
Am J Kidney Dis, 67 (2016), pp. 787-791
[52]
G.A. Herrera.
Proximal tubulopathies associated with monoclonal light chains: the spectrum of clinicopathologic manifestations and molecular pathogenesis.
Arch Pathol Lab Med, 138 (2014), pp. 1365-1380
[53]
T. Messiaen, S. Deret, B. Mougenot, F. Bridoux, P. Dequiedt, J.J. Dion, et al.
Adult Fanconi syndrome secondary to light chain gammopathy clinicopathologic heterogeneity and unusual features in 11 patients.
Medicine (Baltimore), 79 (2000), pp. 135-154
[54]
F. Bridoux, C. Sirac, V. Hugue, C. Decourt, A. Thierry, N. Quellard, et al.
Fanconi's syndrome induced by a monoclonal Vkappa3 light chain in Waldenstrom's macroglobulinemia.
Am J Kidney Dis, 45 (2005), pp. 749-757
[55]
M.B. Stokes, B. Aronoff, D. Siegel, V.D. D’Agati.
Dysproteinemia-related nephropathy associated with crystal-storing histiocytosis.
Kidney Int, 70 (2006), pp. 597-602
[56]
C. El Hamel, A. Thierry, P. Trouillas, F. Bridoux, C. Carrion, N. Quellard, et al.
Crystal-storing histiocytosis with renal Fanconi syndrome: pathological and molecular characteristics compared with classical myeloma-associated Fanconi syndrome.
Nephrol Dial Transplant, 25 (2010), pp. 2982-2990
[57]
C.P. Larsen, J.M. Bell, A.A. Harris, N.C. Messias, Y.H. Wang, P.D. Walker.
The morphologic spectrum and clinical significance of light chain proximal tubulopathy with and without crystal formation.
Mod Pathol, 24 (2011), pp. 1462-1469
[58]
V. Gupta, M. El Ters, K. Kashani, N. Leung, S.H. Nasr.
Crystalglobulin-induced nephropathy.
J Am Soc Nephrol, 26 (2015), pp. 525-529
[59]
T. Tsuji, Y. Itoh, T. Nakamura, Y. Toyozumi, N. Arima, H. Tsuda.
Crystalglobulinemia syndrome due to monoclonal gammopathy of renal significance.
QJM, 108 (2015), pp. 417-418
[60]
X. Gu, G.A. Herrera.
Light-chain-mediated acute tubular interstitial nephritis: a poorly recognized pattern of renal disease in patients with plasma cell dyscrasia.
Arch Pathol Lab Med, 130 (2006), pp. 165-169
[61]
J. Lin, G.S. Markowitz, A.M. Valeri, N. Kambham, W.H. Sherman, G.B. Appel, et al.
Renal monoclonal immunoglobulin deposition disease: the disease spectrum.
J Am Soc Nephrol, 12 (2001), pp. 1482-1492
[62]
C. Pozzi, M. D’Amico, G.B. Fogazzi, S. Curioni, F. Ferrario, S. Pasquali, et al.
Light chain deposition disease with renal involvement: clinical characteristics and prognostic factors.
Am J Kidney Dis, 42 (2003), pp. 1154-1163
[63]
S.H. Nasr, A.M. Valeri, L.D. Cornell, M.E. Fidler, S. Sethi, V.D. D’Agati, et al.
Renal monoclonal immunoglobulin deposition disease: a report of 64 patients from a single institution.
Clin J Am Soc Nephrol, 7 (2012), pp. 231-239
[64]
S.H. Nasr, G.S. Markowitz, M.B. Stokes, S.V. Seshan, E. Valderrama, G.B. Appel, et al.
Proliferative glomerulonephritis with monoclonal IgG deposits: a distinct entity mimicking immune-complex glomerulonephritis.
[65]
S.M. Soares, D.J. Lager, N. Leung, E.N. Haugen, F.C. Fervenza.
A proliferative glomerulonephritis secondary to a monoclonal IgA.
Am J Kidney Dis, 47 (2006), pp. 342-349
[66]
S.H. Nasr, A. Satoskar, G.S. Markowitz, A.M. Valeri, G.B. Appel, M.B. Stokes, et al.
Proliferative glomerulonephritis with monoclonal IgG deposits.
J Am Soc Nephrol, 20 (2009), pp. 2055-2064
[67]
R. Masai, H. Wakui, A. Komatsuda, M. Togashi, N. Maki, H. Ohtani, et al.
Characteristics of proliferative glomerulo-nephritis with monoclonal IgG deposits associated with membranoproliferative features.
Clin Nephrol, 72 (2009), pp. 46-54
[68]
E. Guiard, A. Karras, E. Plaisier, J.P. Duong van Huyen, F. Fakhouri, J.P. Rougier, et al.
Patterns of noncryoglobulinemic glomerulonephritis with monoclonal Ig deposits: correlation with IgG subclass and response to rituximab.
Clin J Am Soc Nephrol, 6 (2011), pp. 1609-1616
[69]
S. Sethi, S.V. Rajkumar.
Monoclonal gammopathy-associated proliferative glomerulonephritis.
Mayo Clin Proc, 88 (2013), pp. 1284-1293
[70]
F. Bridoux, E. Desport, V. Frémeaux-Bacchi, C.F. Chong, J.M. Gombert, C. Lacombe, et al.
Glomerulonephritis with isolated C3 deposits and monoclonal gammopathy: a fortuitous association.
Clin J Am Soc Nephrol, 6 (2011), pp. 2165-2174
[71]
L. Zand, A. Kattah, F.C. Fervenza, R.J. Smith, S.H. Nasr, Y. Zhang, et al.
C3 glomerulonephritis associated with monoclonal gammopathy: a case series.
Am J Kidney Dis, 62 (2013), pp. 506-514
[72]
S. Sethi, W.R. Sukov, Y. Zhang, F.C. Fervenza, D.J. Lager, D.V. Miller, et al.
Dense deposit disease associated with monoclonal gammopathy of undetermined significance.
Am J Kidney Dis, 56 (2010), pp. 977-982
[73]
T.S. Jokiranta, A. Solomon, M.K. Pangburn, P.F. Zipfel, S. Meri.
Nephritogenic lambda light chain dimer: a unique human miniautoantibody against complement factor H.
J Immunol, 163 (1999), pp. 4590-4596
[74]
S. Meri, V. Koistinen, A. Miettinen, T. Törnroth, I.J. Seppälä.
Activation of the alternative pathway of complement by monoclonal lambda light chains in membranoproliferative glomerulonephritis.
J Exp Med, 175 (1992), pp. 939-950
[75]
H. Debiec, M. Hanoy, A. Francois, D. Guerrot, S. Ferlicot, C. Johanet, et al.
Recurrent membranous nephropathy in an allograft caused by IgG3 targeting the PLA2 receptor.
J Am Soc Nephrol, 23 (2012), pp. 1949-1954
[76]
C.P. Larsen, J.M. Ambuzs, S.M. Bonsib, C.L. Boils, L.N. Cossey, N.C. Messias, et al.
Membranous-like glomerulopathy with masked IgG kappa deposits.
Kidney Int, 86 (2014), pp. 154-161
[77]
D. Dingli, D.R. Larson, M.F. Plevak, J.P. Grande, R.A. Kyle.
Focal and segmental glomerulosclerosis and plasma cell proliferative disorders.
Am J Kidney Dis, 46 (2005), pp. 278-282
[78]
F. Grundmann, M. Witthus, H. Göbel, T. Kisner, R. Siewert, T. Benzing, et al.
Monoclonal gammopathy-associated pauci-immune extracapillary-proliferative glomerulonephritis successfully treated with bortezomib.
Clin Kidney J, 6 (2013), pp. 327-329
[79]
A. Ali, L. Schlanger, S.H. Nasr, S. Sethi, S.M. Gorbatkin.
Proliferative C4 dense deposit disease, acute thrombotic microangiopathy, a monoclonal gammopathy, and acute kidney failure.
Am J Kidney Dis, 67 (2016), pp. 479-482
[80]
C. Rigothier, Y. Delmas, L.T. Roumenina, C. Contin-Bordes, S. Lepreux, F. Bridoux, et al.
Distal angiopathy and atypical hemolytic uremic syndrome: clinical and functional properties of an anti-factor H IgA antibody.
Am J Kidney Dis, 66 (2015), pp. 331-336
[81]
W. Cheungpasitporn, N. Leung, S. Sethi, M.A. Gertz, F.C. Fervenza.
Refractory atypical hemolytic uremic syndrome with monoclonal gammopathy responsive to bortezomib-based therapy.
Clin Nephrol, 83 (2015), pp. 363-369
[82]
H. Yao, M. Monge, M. Renou, C. Lecaque, M. Jauréguy, C. Presne, et al.
Thrombotic thrombocytopenic purpura due to anti-ADAMTS13 antibodies in multiple myeloma.
Clin Nephrol, 81 (2014), pp. 210-215
[83]
T. Koga, S. Yamasaki, H. Nakamura, A. Kawakami, A. Furusu, T. Taguchi, et al.
Renal thrombotic microangiopathies/thrombotic thrombocytopenic purpura in a patient with primary Sjögren's syndrome complicated with IgM monoclonal gammopathy of undetermined significance.
Rheumatol Int, 33 (2013), pp. 227-230
[84]
M. Doshi, A. Lahoti, F.R. Danesh, V. Batuman, P.W. Sanders.
Paraprotein-related kidney disease: kidney injury from paraproteins—what determines the site of injury?.
Clin J Am Soc Nephrol, 11 (2016), pp. 2288-2294
[85]
C.E. Alpers, J. Kowalewska.
Fibrillary glomerulonephritis and immunotactoid glomerulopathy.
J Am Soc Nephrol, 19 (2008), pp. 34-37
[86]
M.H. Rosner, A. Edeani, M. Yanagita, I.G. Glezerman, N. Leung.
Paraprotein-related kidney disease: diagnosing and treating monoclonal gammopathy of renal significance.
Clin J Am Soc Nephrol, 11 (2016), pp. 2280-2287
[87]
I. Bancu, L. Cañas, F.J. Juega, M. Pérez, S. Malumbres, J. Bonet, et al.
Outcomes of monoclonal gammopathy of undetermined significance in patients who underwent kidney transplantation.
Transplant Proc, 47 (2015), pp. 2344-2345
[88]
T.E. Goebel, N.K. Schiltz, K.J. Woodside, A.C. Pillai, P.F. Caimi, H.M. Lazarus, et al.
Neoplastic and non-neoplastic complications of solid organ transplantation in patients with preexisting monoclonal gammopathy of undetermined significance.
Clin Transplant, 29 (2015), pp. 851-857
[89]
L. Zand, E.C. Lorenz, F.G. Cosio, F.C. Fervenza, S.H. Nasr, M.J. Gandhi, et al.
Clinical findings, pathology, and outcomes of C3GN after kidney transplantation.
J Am Soc Nephrol, 25 (2014), pp. 1110-1117
[90]
S.H. Nasr, S. Sethi, L.D. Cornell, M.E. Fidler, M. Boelkins, F.C. Fervenza, et al.
Proliferative glomerulonephritis with monoclonal IgG deposits recurs in the allograft.
Clin J Am Soc Nephrol, 6 (2011), pp. 122-132
[91]
N. Leung, D.J. Lager, M.A. Gertz, K. Wilson, S. Kanakiriya, F.C. Fervenza.
Long-term outcome of renal transplantation in light-chain deposition disease.
Am J Kidney Dis, 43 (2004), pp. 147-153
[92]
P.G. Czarnecki, D.J. Lager, N. Leung, A. Dispenzieri, F.G. Cosio, F.C. Fervenza.
Long-term outcome of kidney transplantation in patients with fibrillary glomerulonephritis or monoclonal gammopathy with fibrillary deposits.
Kidney Int, 75 (2009), pp. 420-427
[93]
L.M. Dember.
Amyloidosis-associated kidney disease.
J Am Soc Nephrol, 17 (2006), pp. 3458-3471
[94]
A. Dispenzieri, R.A. Kyle, M.Q. Lacy, S.V. Rajkumar, T.M. Therneau, D.R. Larson, et al.
POEMS syndrome: definitions and long-term outcome.
Blood, 101 (2003), pp. 2496-2506
[95]
F. Rongioletti, A. Rebora.
Updated classification of papular mucinosis, lichen myxedematosus, and scleromyxedema.
J Am Acad Dermatol, 44 (2001), pp. 273-281
[96]
N. Leung, D.R. Barnidge, C.A. Hutchison.
Laboratory testing in monoclonal gammopathy of renal significance (MGRS).
Clin Chem Lab Med, 54 (2016), pp. 929-937
[97]
N. Leung, M. Gertz, R.A. Kyle, F.C. Fervenza, M.V. Irazabal, A. Eirin, et al.
Urinary albumin excretion patterns of patients with cast nephropathy and other monoclonal gammopathy-related kidney diseases.
Clin J Am Soc Nephrol, 7 (2012), pp. 1964-1968
[98]
J.A. Katzmann, A. Dispenzieri, R.A. Kyle, M.R. Snyder, M.F. Plevak, D.R. Larson, et al.
Elimination of the need for urine studies in the screening algorithm for monoclonal gammopathies by using serum immunofixation and free light chain assays.
Mayo Clin Proc, 81 (2006), pp. 1575-1578
[99]
M. Rao, J.L. Lamont, J. Chan, T.W. Concannon, R. Comenzo, S.J. Ratichek, et al.
Serum free light chain analysis for the diagnosis, management, and prognosis of plasma cell dyscrasias: future research needs: identification of future research needs from comparative effectiveness review N.° 73.
Agency for Healthcare Research and Quality (US), (2012),
Report No.: 12-EHC135-EF. AHRQ Future Research Needs Papers
[100]
E. Jenner.
Serum free light chains in clinical laboratory diagnostics.
Clin Chim Acta, 427 (2014), pp. 15-20
[101]
G. Palladini, P. Russo, T. Bosoni, L. Verga, G. Sarais, F. Lavatelli, et al.
Identification of amyloidogenic light chains requires the combination of serum-free light chain assay with immunofixation of serum and urine.
Clin Chem, 55 (2009), pp. 499-504
[102]
C.A. Hutchison, S. Harding, P. Hewins, G.P. Mead, J. Townsend, A.R. Bradwell, et al.
Quantitative assessment of serum and urinary polyclonal free light chains in patients with chronic kidney disease.
Clin J Am Soc Nephrol, 3 (2008), pp. 1684-1690
[103]
C.A. Hutchison, T. Plant, M. Drayson, P. Cockwell, M. Kountouri, K. Basnayake, et al.
Serum free light chain measurement aids the diagnosis of myeloma in patients with severe renal failure.
BMC Nephrol, 9 (2008), pp. 11
[104]
G. Palladini, A. Dispenzieri, M.A. Gertz, S. Kumar, A. Wechalekar, P.N. Hawkins, 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, 30 (2012), pp. 4541-4549
[105]
J.R. Tate, D. Gill, R. Cobcroft, P.E. Hickman.
Practical considerations for the measurement of free light chains in serum.
Clin Chem, 49 (2003), pp. 1252-1257
[106]
M. Ramirez-Alvarado, C.J. Ward, B.Q. Huang, X. Gong, M.C. Hogan, B.J. Madden, et al.
Differences in immunoglobulin light chain species found in urinary exosomes in light chain amyloidosis (AL).
[107]
J.P. Fermand, F. Bridoux, R.A. Kyle, E. Kastritis, B.M. Weiss, M.A. Cook, International Kidney and Monoclonal Gammopathy Research Group.
How I treat monoclonal gammopathy of renal significance (MGRS).
Blood, 122 (2013), pp. 3583-3590
[108]
A.D. Wechalekar, J.D. Gillmore, P.N. Hawkins.
Systemic amyloidosis.
Lancet, 387 (2016), pp. 2641-2654
[109]
C.P. Larsen, C.L. Boils, L.N. Cossey, S.G. Sharma, P.D. Walker.
Clinicopathologic features of membranous-like glomerulopathy with masked IgG kappa deposits.
Kidney Int Rep, 1 (2016), pp. 299-305
[110]
V. Javaugue, A. Karras, F. Glowacki, B. McGregor, C. Lacombe, J.M. Goujon, et al.
Long-term kidney disease outcomes in fibrillary glomerulonephritis: a case series of 27 patients.
Am J Kidney Dis, 62 (2013), pp. 679-690
[111]
J.J. Hogan, B.M. Weiss.
Bridging the divide: an onco-nephrologic approach to the monoclonal gammopathies of renal significance.
Clin J Am Soc Nephrol, 11 (2016), pp. 1681-1691
[112]
S.S. Motwani, L. Herlitz, D. Monga, K.D. Jhaveri, A.Q. Lam.
Paraprotein-related kidney disease: glomerular diseases associated with paraproteinemias.
Clin J Am Soc Nephro, 11 (2016), pp. 2260-2272
[113]
A.A. Chanan-Khan, J.L. Kaufman, J. Mehta, P.G. Richardson, K.C. Miller, S. Lonial, et al.
Activity and safety of bortezomib in multiple myeloma patients with advanced renal failure: a multicenter retrospective study.
Blood, 109 (2007), pp. 2604-2606
[114]
J.F. San-Miguel, P.G. Richardson, P. Sonneveld, M.W. Schuster, D. Irwin, E.A. Stadtmauer, et al.
Efficacy and safety of bortezomib in patients with renal impairment: results from the APEX phase 3 study.
Leukemia, 22 (2008), pp. 842-849
[115]
S. Meister, U. Schubert, K. Neubert, K. Herrmann, R. Burger, M. Gramatzki, et al.
Extensive immunoglobulin production sensitizes myeloma cells for proteasome inhibition.
Cancer Res, 67 (2007), pp. 1783-1792
[116]
G. Bianchi, L. Oliva, P. Cascio, N. Pengo, F. Fontana, F. Cerruti, et al.
The proteasome load versus capacity balance determines apoptotic sensitivity of multiple myeloma cells to proteasome inhibition.
Blood, 113 (2009), pp. 3040-3049
[117]
H. Ludwig, J. Drach, H. Graf, A. Lang, J.G. Meran.
Reversal of acute renal failure by bortezomib-based chemotherapy in patients with multiple myeloma.
Haematologica, 92 (2007), pp. 1411-1414
[118]
P. Moreau, H. Pylypenko, S. Grosicki, I. Karamanesht, X. Leleu, M. Grishunina, et al.
Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study.
Lancet Oncol, 12 (2011), pp. 431-440
[119]
S. Knop, C. Straka, M. Haen, R. Schwedes, H. Hebart, H. Einsele.
The efficacy and toxicity of bendamustine in recurrent multiple myeloma after high-dose chemotherapy.
Haematologica, 90 (2005), pp. 1287-1288
[120]
W. Pönisch, M. Andrea, I. Wagner, D. Hammerschmidt, U. Kreibich, A. Schwarzer, 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, 138 (2012), pp. 1405-1412
[121]
M. Rummel, U. Kaiser, C. Balser, M. Stauch, W. Brugger, M. Welslau, et al.
Bendamustine plus rituximab versus fludarabine plus rituximab for patientswith relapsed indolent and mantle-cell lymphomas: a multicentre, randomised, open-label, non-inferiority phase 3 trial.
Lancet Oncol, 17 (2016), pp. 57-66
[122]
A. Badros, B. Barlogie, E. Siegel, J. Roberts, C. Langmaid, M. Zangari, et al.
Results of autologous stem cell transplant in multiple myeloma patients with renal failure.
Br J Haematol, 114 (2001), pp. 822-829
[123]
M.V. Irazabal, A. Eirin, M.A. Gertz, A. Dispenzieri, S. Kumar, F.K. Buadi, et al.
Acute kidney injury during leukocyte engraftment after autologous stem cell transplantation in patients with light-chain amyloidosis.
Am J Hematol, 87 (2012), pp. 51-54
[124]
M.A. Gertz, D. Dingli.
How we manage autologous stem cell transplantation for patients with multiple myeloma.
[125]
M.A. Dimopoulos, A. Oriol, H. Nahi, J. San-Miguel, N.J. Bahlis, S.Z. Usmani, et al.
Daratumumab, lenalidomide and dexamethasone for multiple myeloma.
N Engl J Med, 375 (2016), pp. 1319-1331
[126]
N. Chen, H. Lau, L. Kong, G. Kumar, J.B. Zeldis, R. Knight, et al.
Pharmacokinetics of lenalidomide in subjects with various degrees of renal impairment and in subjects on hemodialysis.
J Clin Pharmacol, 47 (2007), pp. 1466-1475
[127]
R. Niesvizky, T. Naib, P.J. Christos, D. Jayabalan, J.R. Furst, J. Jalbrzikowski, et al.
Lenalidomide-induced myelosuppression is associated with renal dysfunction: adverse events evaluation of treatment-naïve patients undergoing front-line lenalidomide and dexamethasone therapy.
Br J Haematol, 138 (2007), pp. 640-643
[128]
R. Specter, V. Sanchorawala, D.C. Seldin, A. Shelton, S. Fennessey, K.T. Finn, et al.
Kidney dysfunction during lenalidomide treatment for AL amyloidosis.
Nephrol Dial Transplant, 26 (2011), pp. 881-886
[129]
E. Harris, J. Behrens, D. Samson, A. Rahemtulla, N.H. Russell, J.L. Byme.
Use of thalidomide in patients with myeloma and renal failure may be associated with unexplained hyperkalaemia.
Br J Haematol, 122 (2003), pp. 160-161
[130]
R. Ramos, R. Poveda, C. Bernís, J. Ara, M. Sunyer, P. Arrizabalaga, et al.
Renal involvement in benign monoclonal gammopathies: an underdiagnosed condition.
Nefrologia, 28 (2008), pp. 525-529
[131]
M. Espinosa, R. Ortega, M. Sánchez, A. Segarra, M.T. Salcedo, F. González, et al.
Spanish Group for Study of Glomerular Diseases (GLOSEN) Association of C4d deposition with clinical outcomes in IgA nephropathy.
Clin J Am Soc Nephrol, 9 (2014), pp. 897-904
[132]
J. Caro, E. Gutiérrez-Solís, J. Rojas-Rivera, I. Agraz, N. Ramos, C. Rabasco, Grupo de Estudio de las Enfermedades Glomerulares de la Sociedad Española de Nefrología (GLOSEN), et al.
Predictors of response and relapse in patients with idiopathic membranous nephropathy treated with tacrolimus.
Nephrol Dial Transplant, 30 (2015), pp. 467-474
[133]
C. Rabasco, T. Cavero, E. Román, J. Rojas-Rivera, T. Olea, M. Espinosa, Spanish Group for the Study of Glomerular Diseases (GLOSEN), et al.
Effectiveness of mycophenolate mofetil in C3 glomerulonephritis.
Kidney Int, 88 (2015), pp. 1153-1160

Please cite this article as: Caravaca-Fontán F, Gutiérrez E, Delgado Lillo R, Praga M. Gammapatías monoclonales de significado renal. Nefrologia. 2017;37:465–477.

Copyright © 2017. Sociedad Española de Nefrología
Idiomas
Nefrología (English Edition)
Article options
Tools
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?