Skip to main content
Erschienen in: memo - Magazine of European Medical Oncology 1/2021

Open Access 06.01.2021 | short review

Monoclonal gammopathy of renal significance

A short clinical approach

verfasst von: Thomas Reiter, MD, Maja Nackenhorst, MD

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 1/2021

Summary

Monoclonal gammopathy of renal significance (MGRS) encompasses a group of kidney disorders in which a monoclonal immunoglobulin secreted by a B cell or plasma cell clone causes renal damage, without meeting hematological criteria for malignancy. The underlying disorder in patients with MGRS is generally consistent with monoclonal gammopathy of undetermined significance (MGUS). Because of the wide spectrum of MGRS-associated diseases, defined through the location and mechanism of renal injury, it is often challenging to establish the right diagnosis. Kidney biopsy must be considered early; hence, close cooperation between hematologist and nephrologists is crucial in diagnosis and treatment from the beginning to prevent irreversible organ damage. Anti B‑cell or plasma-cell clone directed therapy with cytostatic or immunomodulatory agents can save and ameliorate renal function significantly. This is underlined by the fact that, untreated, MGRS-associated disease shows early recurrence in patients after kidney transplantation.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AL
Light chain amyloidosis
cHR
Complete hematological response
CKD
Chronic kidney disease
CKD-EPI
Chronic Kidney Disease—Epidemiology Collaboration
FFPE
Formalin fixed paraffin embedded
GBM
Glomerular basal membrane
GFR
Glomerular filtration rate
GN
Glomerulonephritis
MGUS
Monoclonal gammopathy of undetermined significance
MGRS
Monoclonal gammopathy of renal significance
MIDD
Monoclonal immunoglobulin deposition disease
MM
Multiple myeloma
MPGN
Membranoroliferative glomerulonephritis
PGNMID
Proliferative glomerulonephritis associated with monoclonal immune deposits
POEMS
Polyneuropathy, organomegaly, endocrinopathy, myeloma protein, and skin changes
RCT
Randomized controlled trial
TMA
Thrombotic microangiopathy
VGPR
Very good partial response

Introduction

In three percent of a general population over the age of 50 years, monoclonal gammopathy of undetermined significance (MGUS) can be found [1]. MGUS is defined as a premalignant condition, characterized by the presence of a monoclonal immunoglobulin or its fragments < 30 g/l in serum and < 10% of monoclonal bone marrow plasma cells in a patient who does not show any further organ damage attributable to the monoclonal immunoglobulin [2]. The monoclonal immunoglobulins are either products of plasma cells or (small) B cell clones, as they occur in conditions such as multiple myeloma, chronic lymphatic leukemia (CLL), Waldenström’s macroglobulinemia, or low-grade lymphoma.
Progression of MGUS to a hematologic malignancy shows a risk of 1% per year and is indicated by the occurrence of disease-defining events [3]. Multiple myeloma is defined by increase of FLC ratio (involved:uninvolved serum free light-chain ratio) ≥ 100, an increase to monoclonal bone marrow plasma cells ≥ 60% or more than one lytic bone lesion over 5 mm in diameter on magnetic resonance imaging (MRI) [3]. All criteria of differentiation between a premalignant or malignant state include any form of tumor or cell burden.
However, a wide spectrum of renal injuries is associated with the presence of monoclonal immunoglobulins, aside from meeting criteria of hematologic malignancy. This indicates paraprotein-associated nephrotoxicity, independent from the serum-concentration of the toxic monoclonal protein is causal for the renal injury.
Taking this into account, in 2012, the International Kidney and Monoclonal Gammopathy Research Group (IKMG) introduced the term of monoclonal gammopathy of renal significance (MGRS) [4].
In a recent consensus statement, the definition was further narrowed and defined. The term MGRS now applies specifically to any clonal B‑cell lymphoproliferative or clonal plasma-cell proliferative disease with two characteristics: First, one or more kidney lesions must be related to the (produced) monoclonal immunoglobulin. Second, the underlying B cell or plasma cell clone must not be detectable in peripheral blood or bone marrow. The disease-initiating paraprotein can be produced by a localized clonal disease-manifestation as localized plasmacytoma or localized lymph nodes in low grade non-Hodgkin lymphoma (NHL). Therefore not only blood, urine and bone marrow analses should be performed but also whole-body examination with CT scan or PET-CT scan should be considered [5].

When and why to think of MGRS

The incidence of MGUS increases with age (3% in patients > 50 years, 5% in patients aged > 70 years and up to 8% in patients aged 80 years or more) [5]. The incidence of chronic kidney disease (CKD) also increases up to 3.5% in Austria after the age of 60 years [6] and physiologically, glomerular filtration rate decreases after reaching its peak at the age of about 20 years. Therefore, a patient can have CKD and MGUS that are completely unrelated. It is crucial that nephrologists and hematologists cooperate closely to determine whether a renal impairment is caused by MGRS. The importance of this cooperation is underlined by the fact that patients with untreated MGRS are at high risk of developing end stage renal disease with dependence on maintenance dialysis or even the need of kidney transplantation. In a study of 19 patients with monoclonal immunoglobulin deposition disease (MIDD), an MGRS entity, renal survival was only 37% despite overall survival in this cohort being 70% [7]. Maintenance dialysis had worse unadjusted 5‑year survival (50.8%; 95% confidence interval [CI] 50.1–51.6%) compared with prostate (83.3%) and colorectal (56.1%) cancer in men and worse unadjusted 5‑year survival (49.8%) compared with breast (82.1%) and colorectal (56.8%) cancer in women [8]. Further, untreated MGRS has a high rate of recurrence in transplant recipients and can lead to rapid graft loss [9]. Therefore, any reluctance of treatment initiation critically reduces the patient’s survival.

Spectrum of disease and clinical manifestations

MGRS encompasses a multitude of pathologies and clinical manifestations (Table 1). Regarding the location of the lesion in the kidney, the clinical presentation of MGRS may include a rapid decline in glomerular filtration rate and function by presence of proteinuria and/or albuminuria in small or even nephrotic range, hematuria or glucose and serum electrolyte disorders, indicating Fanconi syndrome cause by tubular injury. The pathologic spectrum of MGRS-associated disorders can mimic an otherwise classified renal diseases.
Table 1
MGRS-associated diseases. (Modified from [5])
Monoclonal immunoglobulin deposits
Organized
Fibrillar
Microtubular
Inclusions/crystalline deposits
Immunoglobulin-related Amyloidosis
Immunotactoid GN
LCPT
Monoclonal fibrillary GN
Cryoglobulinaemic GN type I & II
Crystal storing histiocytosis
(Cryo) crystalglobulin GN
Non-organized
MIDD
Proliferative glomerulonephritis associated with monoclonal immune deposits (PGNMID)
No monoclonal immunoglobulin deposits
C3 glomerulopathy with monoclonal gammopathy
Thrombotic microangiopathy
GN glomerulonephritis, LCPT light chain proximal tubulopathy
The most common renal manifestations in MGRS are renal light-chain amyloidosis (AL amyloidosis), monoclonal immunoglobulin deposition disease (MIDD) and cryoglobulinemic glomerulonephritis. Nevertheless, the monoclonal immunoglobulin can also work as a fully functional antibody and cause otherwise unrelated renal manifestations. For example, the monoclonal immunoglobulin can mimic antibodies directed against the renal glomerular basal membrane (GBM). In this case the patient would clinically present with Goodpasture syndrome, although regular antibodies against GBM could not be found with conventional assays [10]. As described in several case series monoclonal immunoglobulins can also act as antibodies against complement regulators and consequently cause thrombotic microangiopathy (TMA) [11].
In both of the examples above the determination of the underlying cell clone can be challenging. Hence, MGRS must be considered in all autoimmune renal disease as a rule-out diagnosis. A previously stated diagnosis must be re-evaluated, especially in clinical courses refractory to standard immunosuppression.
Untreated, MGRS-associated renal disease does unfortunately show early recurrence (60–89%) after kidney transplantation and leads to rapid loss of allograft function [12, 13].

Diagnostic approach

Although most patients will present with symptoms of renal impairment, with proteinuria (> 300 mg/day) and with or without hematuria, all patients with MGUS should be screened on a regular (at least every 3 months) basis for the following parameters:
  • Spot urine examination to determine proteinuria, albuminuria and hematuria, spot urine electrolytes.
  • Serum creatinine and estimation of glomerular filtration rate (CKD-EPI)
  • Serum electrolytes and glucose levels
If not previously performed, the lab work also includes serological and urine electrophoresis, immunofixation in serum and urine, and measurement of free light chain concentration in the serum and 24 h urine collection. Furthermore, a urinary sediment should be evaluated for hematuria or signs of nephritic changes. Immunofixation in 24 h urine collection can be pivotal to determine the underlying clone. In contrast, the urinary free light chain concentration in spot urine does not have any known value with regard to these disorders. For correct diagnosis and monitoring of a monoclonal paraprotein the free light chain concentration in 24 h urine collection is of pivotal importance.
Bone marrow biopsy for hematologic analysis and further whole-body examinations should be performed in accordance with the hematologists.
If the development of rapid deterioration of kidney function indicated by a decline of eGFR of > 2 ml/min/1.73 m2 per year and the patient has a preknown MGUS, MGRS-associated disease must be suspected. These patients have to undergo kidney biopsy. This is the gold standard for diagnosis, and the complex histological workup will give guidance for the further treatment. Complications of kidney biopsies, such as major bleeding, are rare and compatible with those of renal transplant biopsies (4.0 and 3.3% respectively, p = 0.4) (Table 2; [14, 15]).
Table 2
Indications for renal biopsy. (Modified from [5])
 
CKD stage
eGFR (CKD-EPI)
Urine findings
Other findings
Biopsy advised
3 and higher
< 60 ml/min/1.73 m2
&
decline > 2 ml/min/1.73 m2/year
Proteinuria, hematuria
Albumin:creatinine ratio > 300 mg/dl
Fanconi syndrome (hypouricemia)
Biopsy considered
1.2
< 60 ml/min/1.73 m2
Albumine:creatinine ratio 30–300 m mg/dL
Hematuria
Evidence of light chain proteinuria
eGFR estimated glomerular filtration rate, CKD-EPI Chronic Kidney Disease Epidemiology Collaboration, CKD chronic kidney diseases

Histological findings

While clinical manifestations are associated with the renal location of the lesion, the systematic classification of MGRS is guided by the type of organization of monoclonal immunoglobulins in the lesion. Basically MGRS lesions are divided into organized and nonorganized patterns with several subdivisions and further into non-immunoglobulin deposit conditions (Table 1). MGRS has an abundance of light-microscopic and ultrastructural manifestations that can be challenging to interpret [16].
Light microscopy:
For a first overview 2–3 μm sections from formalin fixed and paraffin embedded (FFPE) material are stained with HE (hematoxilin-eosin), PAS, a silver stain (see Fig. 1) and a trichrome stain. This allows a first assessment of predominant pathologies, like deposition of matrix suspicious for amyloidosis in glomeruli, blood vessels and/or the interstitium; thickening of the intraglomerular capillary walls potentially indicating immune-complex or light chain deposition; “hyaline” thrombi in glomerular capillaries as seen in cryoglobulinemia; or tubular casts suspicious for cast nephropathy. For the diagnosis of amyloid deposition a positive Congo red stain (apple-green birefringence under polarized light) performed on a 5–6 μm thick section from FFPE material is mandatory and is at times essential to detect small deposits that can be easily overlooked in other stains [17, 18].
Immunohistochemistry/Immunofluorescence:
Multiple MGRS-associated pathologies show immunohistochemical abnormalities (light chain restrictions in cast nephropathy and AL amyloidosis, LCDD (Light Chain Deposition Disease), C3 deposits in C3 glomerulopathies, etc.). It is imperative to distinguish these entities by thorough immunohistochemical analysis with staining for IgG, IgM, IgA, C3 and C1q as well as kappa and lambda light chains to identify monotypic deposits. This can be performed by immunohistochemistry on FFPE material with the advantage of colocalizing deposition of immunoglobulins and/or complement factors to regions of interest seen in histological stains. Alternatively, as it is custom in several countries, a second core or part of the renal biopsy can be used unfixed for direct or indirect immunofluorescence on frozen sections. This method may be beneficial in case of high background sometimes seen in sections from FFPE material and thus higher specificity. (see Fig. 1)
Ultrastructural analysis:
The last step to define the MGRS manifestation is the ultrastructural analysis. Electron microscopy can confirm light chain deposition disease and help to differentiate between intramembranous, subepithelial and subendothelial deposits, but also identifies mesangial deposits. Further, it can define the structure of deposits seen: Fibrillary deposits are found in Ig-related amyloidosis and fibrillary glomerulonephritis (GN), microtubules are found in immunotactoid GN and can be found in type 1 cryoglobulinemic GN [19, 20]. Crystal formation can be found in light chain proximal tubulopathy and crystal storing histocytosis [21, 22]. Nonorganized deposits are found in the monoclonal immunoglobulin deposition diseases (MIDD), PGNMID and C3 glomerulopathy, with associated monoclonal gammopathy. C3 glomerulopathy encompasses dense deposit disease (DDD) with prominent intramembranous and to a lesser extent mesangial deposits, as well as C3 glomerulonephritis with less well defined mesangial, intramembranous and subendothelial deposits [23].

Treatment

After diagnosis of an MGRS-associated disease, therapy should be initiated immediately. The goal of MGRS therapy is to eliminate the underlying clone, while preserving or ameliorating kidney function. The clonal-directed treatment should be managed by hematologists and nephrologists in close cooperation. For the management of these complex diseases interdisciplinary cooperation between nephrologists, hematologists, specialized nephropathologists, radiologists and transplant specialists is of utmost importance. Thus, diagnosis and treatment should be performed in specialized centers with high level of experience.
Take home message
Untreated, MGRS leads to end stage renal disease and unfortunately, to rapid loss of graft function in patients after kidney transplantation. Interdisciplinary cooperation is crucial for renal and overall survival.

Conflict of interest

T. Reiter and M. Nackenhorst declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Kyle RA, Therneau TM, Rajkumar SV, Larson DR, Plevak MF, Offord JR, et al. Prevalence of monoclonal gammopathy of undetermined significance. N Engl J Med. 2006;354(13):1362–9.CrossRef Kyle RA, Therneau TM, Rajkumar SV, Larson DR, Plevak MF, Offord JR, et al. Prevalence of monoclonal gammopathy of undetermined significance. N Engl J Med. 2006;354(13):1362–9.CrossRef
2.
Zurück zum Zitat Kyle RA. Monoclonal gammopathy of undetermined significance. Natural history in 241 cases. Am J Med. 1978;64(5):814–26.CrossRef Kyle RA. Monoclonal gammopathy of undetermined significance. Natural history in 241 cases. Am J Med. 1978;64(5):814–26.CrossRef
3.
Zurück zum Zitat Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, Mateos MV, et al. International myeloma working group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15(12):e538–e48.CrossRef Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, Mateos MV, et al. International myeloma working group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15(12):e538–e48.CrossRef
4.
Zurück zum Zitat Leung N, Bridoux F, Hutchison CA, Nasr SH, Cockwell P, Fermand JP, et al. Monoclonal gammopathy of renal significance: when MGUS is no longer undetermined or insignificant. Blood. 2012;120(22):4292–5.CrossRef Leung N, Bridoux F, Hutchison CA, Nasr SH, Cockwell P, Fermand JP, et al. Monoclonal gammopathy of renal significance: when MGUS is no longer undetermined or insignificant. Blood. 2012;120(22):4292–5.CrossRef
5.
Zurück zum Zitat Leung N, Bridoux F, Batuman V, Chaidos A, Cockwell P, D’Agati VD, et al. The evaluation of monoclonal gammopathy of renal significance: a consensus report of the International Kidney and Monoclonal Gammopathy Research Group. Nat Rev Nephrol. 2019;15(1):45–59.CrossRef Leung N, Bridoux F, Batuman V, Chaidos A, Cockwell P, D’Agati VD, et al. The evaluation of monoclonal gammopathy of renal significance: a consensus report of the International Kidney and Monoclonal Gammopathy Research Group. Nat Rev Nephrol. 2019;15(1):45–59.CrossRef
7.
Zurück zum Zitat Heilman RL, Velosa JA, Holley KE, Offord KP, Kyle RA. Long-term follow-up and response to chemotherapy in patients with light-chain deposition disease. Am J Kidney Dis. 1992;20(1):34–41.CrossRef Heilman RL, Velosa JA, Holley KE, Offord KP, Kyle RA. Long-term follow-up and response to chemotherapy in patients with light-chain deposition disease. Am J Kidney Dis. 1992;20(1):34–41.CrossRef
8.
Zurück zum Zitat Naylor KL, Kim SJ, McArthur E, Garg AX, McCallum MK, Knoll GA. Mortality in incident maintenance dialysis patients versus incident solid organ cancer patients: a population-based cohort. Am J Kidney Dis. 2019;73(6):765–76.CrossRef Naylor KL, Kim SJ, McArthur E, Garg AX, McCallum MK, Knoll GA. Mortality in incident maintenance dialysis patients versus incident solid organ cancer patients: a population-based cohort. Am J Kidney Dis. 2019;73(6):765–76.CrossRef
9.
Zurück zum Zitat 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;43(1):147–53.CrossRef 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;43(1):147–53.CrossRef
10.
Zurück zum Zitat Borza DB, Chedid MF, Colon S, Lager DJ, Leung N, Fervenza FC. Recurrent Goodpasture’s disease secondary to a monoclonal IgA1-kappa antibody autoreactive with the alpha1/alpha2 chains of type IV collagen. Am J Kidney Dis. 2005;45(2):397–406.CrossRef Borza DB, Chedid MF, Colon S, Lager DJ, Leung N, Fervenza FC. Recurrent Goodpasture’s disease secondary to a monoclonal IgA1-kappa antibody autoreactive with the alpha1/alpha2 chains of type IV collagen. Am J Kidney Dis. 2005;45(2):397–406.CrossRef
11.
Zurück zum Zitat Ravindran A, Go RS, Fervenza FC, Sethi S. Thrombotic microangiopathy associated with monoclonal gammopathy. Kidney Int. 2017;91(3):691–8.CrossRef Ravindran A, Go RS, Fervenza FC, Sethi S. Thrombotic microangiopathy associated with monoclonal gammopathy. Kidney Int. 2017;91(3):691–8.CrossRef
12.
Zurück zum Zitat Czarnecki PG, Lager DJ, Leung N, Dispenzieri A, Cosio FG, Fervenza FC. Long-term outcome of kidney transplantation in patients with fibrillary glomerulonephritis or monoclonal gammopathy with fibrillary deposits. Kidney Int. 2009;75(4):420–7.CrossRef Czarnecki PG, Lager DJ, Leung N, Dispenzieri A, Cosio FG, Fervenza FC. Long-term outcome of kidney transplantation in patients with fibrillary glomerulonephritis or monoclonal gammopathy with fibrillary deposits. Kidney Int. 2009;75(4):420–7.CrossRef
13.
Zurück zum Zitat Herrmann SM, Gertz MA, Stegall MD, Dispenzieri A, Cosio FC, Kumar S, 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;26(6):2032–6.CrossRef Herrmann SM, Gertz MA, Stegall MD, Dispenzieri A, Cosio FC, Kumar S, 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;26(6):2032–6.CrossRef
14.
Zurück zum Zitat Fish R, Pinney J, Jain P, Addison C, Jones C, Jayawardene S, et al. The incidence of major hemorrhagic complications after renal biopsies in patients with monoclonal gammopathies. Clin J Am Soc Nephrol. 2010;5(11):1977–80.CrossRef Fish R, Pinney J, Jain P, Addison C, Jones C, Jayawardene S, et al. The incidence of major hemorrhagic complications after renal biopsies in patients with monoclonal gammopathies. Clin J Am Soc Nephrol. 2010;5(11):1977–80.CrossRef
15.
Zurück zum Zitat Ciocchini M, Musso CG. Why renal biopsy is crucial in monoclonal gammopathy of renal significance (MGRS). Int Urol Nephrol. 2019;51(5):899–900.CrossRef Ciocchini M, Musso CG. Why renal biopsy is crucial in monoclonal gammopathy of renal significance (MGRS). Int Urol Nephrol. 2019;51(5):899–900.CrossRef
16.
Zurück zum Zitat Bridoux F, Leung N, Hutchison CA, Touchard G, Sethi S, Fermand JP, et al. Diagnosis of monoclonal gammopathy of renal significance. Kidney Int. 2015;87(4):698–711.CrossRef Bridoux F, Leung N, Hutchison CA, Touchard G, Sethi S, Fermand JP, et al. Diagnosis of monoclonal gammopathy of renal significance. Kidney Int. 2015;87(4):698–711.CrossRef
17.
Zurück zum Zitat Howie AJ, Brewer DB. Optical properties of amyloid stained by Congo red: history and mechanisms. Micron. 2009;40(3):285–301.CrossRef Howie AJ, Brewer DB. Optical properties of amyloid stained by Congo red: history and mechanisms. Micron. 2009;40(3):285–301.CrossRef
18.
Zurück zum Zitat Picken MM. Modern approaches to the treatment of amyloidosis: the critical importance of early detection in surgical pathology. Adv Anat Pathol. 2013;20(6):424–39.CrossRef Picken MM. Modern approaches to the treatment of amyloidosis: the critical importance of early detection in surgical pathology. Adv Anat Pathol. 2013;20(6):424–39.CrossRef
19.
Zurück zum Zitat Said SM, Sethi S, Valeri AM, Leung N, Cornell LD, Fidler ME, et al. Renal amyloidosis: origin and clinicopathologic correlations of 474 recent cases. Clin J Am Soc Nephrol. 2013;8(9):1515–23.CrossRef Said SM, Sethi S, Valeri AM, Leung N, Cornell LD, Fidler ME, et al. Renal amyloidosis: origin and clinicopathologic correlations of 474 recent cases. Clin J Am Soc Nephrol. 2013;8(9):1515–23.CrossRef
20.
Zurück zum Zitat Bridoux F, Hugue V, Coldefy O, Goujon JM, Bauwens M, Sechet A, et al. Fibrillary glomerulonephritis and immunotactoid (microtubular) glomerulopathy are associated with distinct immunologic features. Kidney Int. 2002;62(5):1764–75.CrossRef Bridoux F, Hugue V, Coldefy O, Goujon JM, Bauwens M, Sechet A, et al. Fibrillary glomerulonephritis and immunotactoid (microtubular) glomerulopathy are associated with distinct immunologic features. Kidney Int. 2002;62(5):1764–75.CrossRef
21.
Zurück zum Zitat Kapur U, Barton K, Fresco R, Leehey DJ, Picken MM. Expanding the pathologic spectrum of immunoglobulin light chain proximal tubulopathy. Arch Pathol Lab Med. 2007;131(9):1368–72.CrossRef Kapur U, Barton K, Fresco R, Leehey DJ, Picken MM. Expanding the pathologic spectrum of immunoglobulin light chain proximal tubulopathy. Arch Pathol Lab Med. 2007;131(9):1368–72.CrossRef
22.
Zurück zum Zitat El Hamel C, Thierry A, Trouillas P, Bridoux F, Carrion C, Quellard N, et al. Crystal-storing histiocytosis with renal Fanconi syndrome: pathological and molecular characteristics compared with classical myeloma-associated Fanconi syndrome. Nephrol Dial Transplant. 2010;25(9):2982–90.CrossRef El Hamel C, Thierry A, Trouillas P, Bridoux F, Carrion C, Quellard N, et al. Crystal-storing histiocytosis with renal Fanconi syndrome: pathological and molecular characteristics compared with classical myeloma-associated Fanconi syndrome. Nephrol Dial Transplant. 2010;25(9):2982–90.CrossRef
23.
Zurück zum Zitat Pickering MC, D’Agati VD, Nester CM, Smith RJ, Haas M, Appel GB, et al. C3 glomerulopathy: consensus report. Kidney Int. 2013;84(6):1079–89.CrossRef Pickering MC, D’Agati VD, Nester CM, Smith RJ, Haas M, Appel GB, et al. C3 glomerulopathy: consensus report. Kidney Int. 2013;84(6):1079–89.CrossRef
Metadaten
Titel
Monoclonal gammopathy of renal significance
A short clinical approach
verfasst von
Thomas Reiter, MD
Maja Nackenhorst, MD
Publikationsdatum
06.01.2021
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 1/2021
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-020-00671-4

Weitere Artikel der Ausgabe 1/2021

memo - Magazine of European Medical Oncology 1/2021 Zur Ausgabe