Sie sind auf Seite 1von 14

http://www.kidney-international.

org review
& 2015 International Society of Nephrology

Diagnosis of monoclonal gammopathy of renal


significance
Frank Bridoux1, Nelson Leung2,3, Colin A. Hutchison4, Guy Touchard1, Sanjeev Sethi5,
Jean-Paul Fermand6, Maria M. Picken7, Guillermo A. Herrera8, Efstathios Kastritis9, Giampaolo Merlini10,
Murielle Roussel11, Fernando C. Fervenza2, Angela Dispenzieri3, Robert A. Kyle3, Samih H. Nasr5
on behalf of the International Kidney and Monoclonal Gammopathy Research Group
1
Department of Nephrology and Transplantation, Centre Hospitalier Universitaire de Poitiers and Centre national de reference maladies
rares: amylose AL et autres maladies a depots dimmunoglobulines monoclonales; Universite de Poitiers, Poitiers, France; 2Division of
Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA; 3Division of Hematology, Department
of Medicine, Mayo Clinic, Rochester, Minnesota, USA; 4Department of Medicine, University of Otago, Wellington, New Zealand;
5
Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA; 6Department of Hematology and
Immunology, Univerisity Hospital St Louis, Paris, France; 7Department of Pathology, Loyola University Medical Center, Maywood, Illinois,
USA; 8Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA; 9Department of Clinical
Therapeutics, Alexandra Hospital, University of Athens School of Medicine, Athens, Greece; 10Department of Molecular Medicine,
University of Pavia, Pavia, Italy and 11Hematologie Clinique, Hopital Purpan, Toulouse, France

Monoclonal gammopathy of renal significance (MGRS) Kidney disease is a frequent complication of monoclonal
regroups all renal disorders caused by a monoclonal gammopathies that manifests with a wide range of renal
immunoglobulin (MIg) secreted by a nonmalignant B-cell lesions. These patterns are mostly determined by the physico-
clone. By definition, patients with MGRS do not meet the chemical characteristics of the pathogenic monoclonal
criteria for overt multiple myeloma/B-cell proliferation, and immunoglobulin (MIg).1 The renal disease may complicate
the hematologic disorder is generally consistent with a previously diagnosed clonal B-cell disorder or be the initial
monoclonal gammopathy of undetermined significance manifestation of the hematological disease. Two main
(MGUS). However, MGRS is associated with high morbidity categories of renal disorders associated with monoclonal
due to the severity of renal and sometimes systemic lesions gammopathies should be distinguished, depending on the
induced by the MIg. Early recognition is crucial, as burden of the underlying plasma cell or B-cell clone. The first
suppression of MIg secretion by chemotherapy often group of renal disorders requires the secretion of large
improves outcomes. The spectrum of renal diseases in MGRS amounts of all or part of the MIg and is only observed in the
is wide, including old entities such as AL amyloidosis and setting of a high tumor mass B-cell proliferation. This is
newly described lesions, particularly proliferative typically illustrated by the massive precipitation of light chain
glomerulonephritis with monoclonal Ig deposits and C3 (LC) in the lumen of distal tubules, which characterizes light-
glomerulopathy with monoclonal gammopathy. Kidney chain cast nephropathy, the most common cause of acute
biopsy is indicated in most cases to determine the exact kidney injury in multiple myeloma (MM). Light-chain cast
lesion associated with MGRS and evaluate its severity. nephropathy is considered as a MM defining event because it
Diagnosis requires integration of morphologic alterations by always forms as the result of high monoclonal LC production,
light microscopy, immunofluorescence (IF), electron indicating a high tumor burden, a situation that requires
microscopy, and in some cases by IF staining for Ig isotypes, urgent introduction of chemotherapy.2,3 Although rare,
immunoelectron microscopy, and proteomic analysis. another example is glomerular intracapillary deposition of
Complete hematologic workup with serum and urine protein MIgM to form thrombi in high tumor mass Waldenstroms
electrophoresis, immunofixation, and serum-free light-chain macroglobulinemia.4
assay is required. This review addresses the pathologic and The other group is renal diseases associated with low-
clinical features of MGRS lesions, indications of renal biopsy, grade lymphoproliferative disorders. Although they may
and a proposed algorithm for the hematologic workup. occur during symptomatic B-cell proliferations, these other
Kidney International advance online publication, 21 January 2015; MIg-related renal lesions are mainly encountered in patients
doi:10.1038/ki.2014.408 with a small B-cell clone and low malignant potential.5 Only
8% of patients with immunoglobulin light-chain (AL)
Correspondence: Nelson Leung, Division of Nephrology and Hypertension, amyloidosis6 and 20% of patients with Randall-type
Mayo Clinic, 200 First Street, Rochester, Minnesota 55905, USA. monoclonal Ig deposition disease (MIDD)7 have evidence
E-mail: leung.nelson@mayo.edu of a symptomatic MM at diagnosis. This clone can be
Received 28 February 2014; revised 17 July 2014; accepted 24 July 2014 expressed as any indolent B-cell lymphoid disorder, including

Kidney International 1
review F Bridoux et al.: MGRS

the so-called smoldering asymptomatic MM and low-grade therapy including stem cell transplantation has been used in
lymphoplasmacytic lymphoma with Waldenstroms macro- AL amyloidosis in which the majority of patients do not meet
globulinemia, or a monoclonal gammopathy of undeter- the criteria for symptomatic MM.11 Hence, this is one of the
mined significance (MGUS). In this setting, structural first examples of successful treatment of a MGRS-related
peculiarities of the MIg, particularly the variable domain, kidney disease. The treatment strategy of MGRS is based on
and not the rate of production, are the main determinants of chemotherapy that should be adapted to the nature of the
renal lesions. The nonmalignant nature of these clones has underlying clone, either lymphocytic or plasmacytic, to
been a source of confusion for clinicians when it comes to renal function and to the presence or not of extrarenal
treatment. Because many of these clones are best classified as involvement.12 Rapid suppression of the secretion of
MGUS, defined as a plasma cell proliferative disorder that nephrotoxic MIg has been shown to favorably impact renal
manifests o3 g/dl of monoclonal protein and o10% bone and patient survival in most diseases associated with
marrow plasma cells, or smoldering MM, defined by 43 g/dl MGRS, including AL amyloidosis, light-chain deposition
of monoclonal protein and/or 410% bone marrow plasma disease (LCDD), immunotactoid glomerulopathy, and proli-
cells but without any end organ damage, chemotherapy is ferative glomerulonephritis with monoclonal Ig deposits
not indicated in patients with MGUS, smoldering MM, or (PGNMID).1316 In AL amyloidosis, renal response is closely
low-grade lymphomas.8 According to the current treatment associated with the magnitude of hematological response, as
guidelines, these patients require only clinical and biological renal response rate is significantly higher in patients who
surveillance as they may remain asymptomatic over a have achieved a serum-free light-chain (FLC) response of
prolonged period of time, and early therapy has not been more than 90%.17 Finally, in the absence of an efficient
shown to be beneficial in the majority of patients. suppression of the nephrotoxic MIg, MGRS-related renal
Chemotherapy is only introduced when symptoms related lesions recur in the allograft in most patients, usually within
to the underlying lymphocytic or plasmacytic proliferative the first year after renal transplantation.1822
process develop or are impending. Symptomatic MM is In addition to AL amyloidosis, the spectrum of MGRS
defined by evidence of end organ damage, characterized by includes various other renal lesions. Most are due to
CRAB (hyperCalcemia, Renal impairment, Anemia, Bone deposition of a MIg fragment with distinct localization and
disease). pattern of ultrastructural organization, usually resulting in
On the basis of the current guidelines, cytotoxic therapy is glomerular disorders but more rarely in tubular disorders
often withheld in patients with renal diseases associated with such as the Fanconi syndrome (FS). Glomerulopathies are
MIg who do not meet the criteria for MM or symptomatic featured either by organized deposits, fibrillar (AL and Ig
lymphoma.9 Such an approach is not appropriate for these heavy chains (AH) amyloidosis) or microtubular (type I
patients, as despite the absence of high tumor burden, they cryoglobulinemic glomerulonephritis, immunotactoid glo-
display high morbidity and even increased mortality. In an merulopathy), or by non-organized deposits (MIDD and
effort to address this need, the International Kidney and PGNMID). Importantly, our understanding of the spectrum
Monoclonal Gammopathy Research Group (IKMG) had of MGRS is evolving, and, in addition to kidney deposition of
their inaugural meeting in Bath, UK, in 2009. Members of a MIg, other mechanisms may be implicated, involving the
the IKMG come from the disciplines of nephrology, secretion of various biological factors and/or autoantibody
hematology, and nephropathology and from countries activity of the MIg. Examples include secretion of vascular
around the globe including Austria, Australia, Canada, endothelial growth factor in the POEMS syndrome (poly-
France, Italy, New Zealand, Spain, United Kingdom, and neuropathy, organomegaly, endocrinopathy, monoclonal
United States. Through a series of subsequent meetings, the gammopathy, and skin changes) that may trigger develop-
term monoclonal gammopathy of renal significance (MGRS) ment of renal endothelial cell injury and thrombotic
was refined and introduced in 2012 to distinguish these microangiopathy-like lesions,23 membranoproliferative
monoclonal gammopathies from MGUS.10 The main goal glomerulonephritis induced by dimeric monoclonal lambda
was to clearly delineate the benign hematological disorder LC acting as a mini-autoantibody against complement
MGUS, which cannot be associated with any end organ factor H,24,25 or membranous nephropathy caused by a
damage, from MGRS that is associated with severe monoclonal IgG3 kappa targeting the phospholipase A2
consequences related to MIg deposition in the kidneys (and receptor.26 Early recognition and prompt characterization of
sometimes in other organs) that considerably increase the type of MGRS is crucial, as it determines the therapeutic
morbidity and may even impair patient survival. It is strategy and strongly impacts renal prognosis.12 The purpose
important to recognize that like MGUS, the diagnosis of of this article is to provide keys for the diagnostic approach
MGRS does not exclude the possibility of future hematologic to recognize MGRS.
disease progression.
With its clear distinction from the benign term of INDICATIONS AND TIMING OF KIDNEY BIOPSY
MGUS, treatment strategies for MGRS-related kidney con- In a patient in whom MGRS is suspected, usually based on
ditions can be formulated without conflict with the current the coexistence of monoclonal gammopathy and renal
MM treatment guidelines. It should be noted that cytotoxic symptoms, it is essential to quickly and accurately assess

2 Kidney International
F Bridoux et al.: MGRS review

the characteristics of the monoclonal gammopathy, the patients with glomerular involvement who display predomi-
underlying B-cell clone, the type of nephropathy, and its nant albuminuria (usually accompanied with secretion of the
impact on renal function. Baseline glomerular filtration rate MIg or a fragment of it) from those with tubulointerstitial
is a major determinant of renal outcome in most types of lesions.37 For instance, in patients with FS, proteinuria typically
MGRS, as shown in AL amyloidosis,17 MIDD,7 and contains a small amount of albumin but predominantly mono-
PGNMID.27 In addition, it is mandatory to carefully search clonal LC and low-molecular weight proteins. These findings
for extrarenal manifestations, which may influence prognosis should prompt search for symptoms of proximal tubule
and influence therapeutic decisions. Rapid diagnostic dysfunction, including normoglycemic glycosuria, uricosuria,
assessment is particularly critical in patients with AL phosphaturia, and proximal renal tubular acidosis.38,39 In those
amyloidosis, in whom concomitant heart, liver, or patients without urine abnormalities but impaired glomerular
peripheral nerve involvement is frequent, results in filtration rate and evidence of monoclonal gammopathy, a
decreased survival, and affects therapeutic strategy. Median kidney biopsy should be performed in the absence of an
survival is approximately 6 months in AL patients with obvious cause of renal disease. Indeed, small cohorts of AL
significant heart involvement.28 Efficient chemotherapy amyloidosis and LCDD patients with predominant vascular/
based on novel agents such as the cyclophosphamide tubulointerstitial deposits and 24 h urine protein excretion of
bortezomibdexamethasone regimen is currently recom- less than 0.5 g/day have been reported.40,41
mended in the presence of amyloid cardiomyopathy to
achieve rapid and deep hematological response, a key factor HEMATOLOGIC EVALUATION
for patient survival.29,30 Severe extrarenal manifestations are The finding of monoclonal Ig deposits in the kidney indicates
also not uncommon in MIDD and type I cryoglobulinemia, the presence of an underlying B-cell clone. Efforts should
and osteomalacia frequently reveals FS. In patients with be made to characterize this clone, which is a key point for
symptoms suggestive of systemic AL amyloidosis, minimally guiding the therapeutic strategy.12 Accordingly, in all cases,
invasive biopsies of abdominal fat and minor salivary glands a detailed hematologic evaluation should be performed
may be performed initially.31,32 (Figure 1). In patients with IgG, IgA, or LC only MGRS,
However, in most situations, a kidney biopsy with detailed bone marrow aspirate, and biopsy are usually sufficient to
immunofluorescence (IF) and electron microscopic (EM) identify the clone. Flow cytometry and/or immunohistolo-
studies to identify deposit composition and pattern of gical studies of bone marrow cells are often useful to detect
organization is needed. Because of the frequency of this clone as it may present without any morphologic
monoclonal gammopathies in patients aged over 50 years, abnormalities. Skeletal radiographs are mandatory to detect
it is important that the correct correlation of the renal lesions bone lesions and can be supplemented by a vertebral (and
with monoclonal gammopathy is performed, as the presence pelvic) magnetic resonance imaging.
of a monoclonal protein by itself is not equal to the causative Particularly, in patients with IgM MGRS, additional
agent. This is illustrated by the fact that up to 10% of patients imaging studies may be required, as the possibility of a
with hereditary amyloidosis were initially misdiagnosed as non-plasmacytic B-cell clone increases. A computer tomo-
AL amyloidosis because of the presence of a serum MIg33 and graphy scan of the chest, abdomen, and pelvis may help
that 10% of patients with amyloidosis derived from leukocyte identify lymph nodes that should undergo a biopsy.
cell-derived chemotaxin 2, which mainly affects the kidneys, The utility of positron emission tomography scan combined
have a serum MIg.34 Moreover, misdiagnosis of the amyloid with a computer tomography scan remains to be assessed.
subtype may occur, because of nonspecific trapping of the Phenotypic characterization of peripheral blood lympho-
MIg in the amyloid deposits.35 In addition, renal biopsy is cytes, including rearrangement of Ig genes, may be indicated.
indicated to assess the MGRS type and evaluate severity of In patients with MGRS, the likelihood of identifying the
renal disease. This remains important even in patients with B-cell clone significantly increases when a serum and/or urine
advanced chronic kidney disease in whom kidney monoclonal protein can be detected.
transplantation is planned, as the disease generally recurs
rapidly in the allograft in the absence of control of the Screening for MIg
underlying clone.1822 Kidney biopsy is a safe procedure in MGRS can present across a number of clinical scenarios,
patients with MGRS, as shown in a recent study of 148 from isolated proteinuria to end-stage kidney disease. It is
patients with monoclonal gammopathies in whom the rate of therefore imperative that the clinician considers the possibil-
hemorrhagic complications was 4.1%, similar to the control ity of underlying monoclonal gammopathy in patients with
population.36 various renal manifestations and performs appropriate tests.
The spectrum of renal manifestations in MGRS is wide and In many cases, the MIg can be identified in the serum or
varies according to disease type and molecular characteristics of urine by a conventional EP. However, in some cases, the MIg
the pathogenic MIg (Tables 13). Renal failure and proteinuria, levels are very small and may not be detected by EP, likely
with or without hematuria, are the most common presenting reflecting the small size of the underlying B-cell clone and/or
symptoms. Electrophoresis (EP) and immunofixation of a 24-h the affinity of the MIg for tissues and organs. In fact, in
urine specimen should be performed in all cases to distinguish several MGRS renal lesions, the MIg may seemingly be

Kidney International 3
review F Bridoux et al.: MGRS

Abbreviations: CKD, chronic kidney disease; IF, immunofluorescence; LC, immunoglobulin light chains; LN, lymph nodes; LPL, lymphoplasmacytic lymphoma; MGRS, monoclonal gammopathy of renal significance; MM, multiple
demonstrated only in a kidney biopsy by IF or immuno-
Hematological disease

Symptomatic MM and
histochemistry.
Serum and urine immunofixation should be performed in
WM uncommon
all cases to identify the MIg isotype, even if the serum protein
EP is not informative, because it is more sensitive for
MGRS

MGRS

MGRS
detecting MIg than serum protein EP. Furthermore, it is more
MM

MM
LPL
sensitive than the FLC assay among patients with small clones
that produce mostly intact immunoglobulins.42,43 Analysis of

Most common symptoms: hypouricemia, hypophosphatemia, normoglycemic glycosuria, generalized aminoaciduria, low-molecular weight proteinuria, and proximal (type 2) renal tubular acidosis.
the serum and urine by western blotting further increases the

lung, skin, cornea


liver, spleen, LN, sensitivity for detecting very low amounts of MIg15 but is not
(osteomalacia)

Bone marrow,
involvement

routinely available. The latter technique allows identification


Extrarenal

of IgG subclass and may detect circulating truncated


None
Bone

monoclonal heavy chain.15


The issue whether serum only screening is a valuable
alternative to assessing both serum and urine was recently
PTC lysosomes or free in the

occasionally in PTC and glo-


Crystals (rhomboid) within

challenged again through the routine use of the FLC assay. In


Increased lysosomes with

Crystals (needle-shaped)
Ultrastructural findings

an assessment of 1877 patients with monoclonal gammopa-


a mottled appearance

within histiocytes and


accumulations of LCs
Amorphous granular

thies, Katzmann et al. found that serum protein EP and a


quantitative serum FLC assay identified 100% of patients with
merular cells

symptomatic MM and Waldenstroms macroglobulinemia,


cytoplasm

99.5% of patients with smoldering MM, 96.5% of patients


with AL amyloidosis, 79% of patients with LCDD, and 89% of
Table 1 | Main clinical, pathological, and immunological characteristics of tubular disorders in MGRS

patients with MGUS.42 The authors concluded that for


optimal sensitivity in AL amyloidosis and LCDD serum and
Mostly kappa :Vk1
Lambda or kappa
PTC LC inclusions

PTC LC inclusions

urine immunofixation was recommended. Similar


PTC LC staining
kappa: Vk1, or
Almost always

conclusions were reached in a study dedicated to the


IF findings

Vk3 (rare)

detection of the monoclonal protein in patients with


(Ig type)

or Vk3

AL amyloidosis, confirming the necessity to perform both


serum and urine immunofixation.43 Although some
international guidelines recommend the combination of
sions (pseudo-pseudo Gaucher cells)

serum EP and FLC measurement alone for the screening of


in the interstitium and perirenal fat
PTC atrophy and dedifferentiation

PTC atrophy and dedifferentiation

PTC atrophy and dedifferentiation

monoclonal gammopathies,44,45 the applicability of this


Histiocytes with crystalline inclu-

strategy must be weighed with considerable caution. As


Intra-cytoplasmic inclusions
Light microscopic findings

PTC cytoplasmic swelling

already discussed, MGRS is frequently associated with a small


clone, which may be difficult to identify, and it may not be
driven by free LC. In addition, the value of urine protein study
varies with the specific MGRS pathology under consideration.
myeloma; PTC, proximal tubular cells; WM, Waldenstroms macroglobulinemia.

Thus, in a case series of 51 individuals with MIDD, Nasr et al.


demonstrated that all had an abnormal serum FLC ratio;
however, only 81% of these had abnormal urinary assessment
using EP and immunofixation.7 In contrast, only 50% of
patients with MPGN secondary to a monoclonal protein had
Proximal tubule dysfunc-

Slowly progressive CKD

an abnormal FLC, whereas 78% and 96% had an abnormal


Tubular proteinuria
Renal symptoms

urine or serum immunofixation, respectively.46 Until robust


progressive CKD

Proximal tubule

data based on large repositories including all types of MGRS


suggest otherwise, we recommend assessment of both serum
dysfunction

and urine in patients suspected to have MGRS.


tiona

CKD

Interpretation of serum FLC assay


To date, the clinical evaluation of FLC assays has been almost
Proximal tubulopathy

Crystal-storing histio-

entirely based on nephelometric immunoassays using poly-


Tubular disorder
Light chain Fanconi

clonal antibodies raised in sheep against LC epitopes, which


without crystals

are exposed when the LCs are free but hidden when the LCs
are bound (Freelite, Binding Site, Birmingham, UK).47 These
syndrome

cytosis

assays, which have high sensitivity, suggest clonality by


comparing the concentration and ratio of kappa to lambda in
a

4 Kidney International
F Bridoux et al.: MGRS review

Abbreviations: AH, immunoglobulin heavy chain; AHL, immunoglobulin heavy and light chain; AL, immunoglobulin light chain; AKI, acute kidney injury; CKD, chronic kidney disease; CLL, chronic lymphocytic leukemia; CW,
glomerular capillary walls; EP, electrophoresis; FLC, serum-free light chain assay; GN, glomerulonephritis; GOMMID, glomerulonephritis with organized microtubular immunoglobulin deposits; HC, immunoglobulin heavy chains;
Hypocomp., hypocomplementemia; IF, immunofluorescence; Ig, immunoglobulin; ITGN, immunotactoid glomerulonephritis; LC, immunoglobulin light chains; MM, multiple myeloma; MPGN, membranoproliferative
Symptomatic MM uncommon
Hematological and immuno-
the serum. In the last few years, new FLC assays based on
monoclonal antibodies have become commercially available.
logical characteristics

They are by definition limited because of the identification of

Hypocomp. common
one epitope on a FLC.4851 In an attempt to overcome this

B-cell lymphoma

B-cell lymphoma
MM uncommon
CLL (common)
limitation, manufacturers frequently use multiple mono-

Hypocomp.
clonal antibodies within one assay batch. Significant work

B30%
now needs to be undertaken to determine the comparability
MGRS

MGRS

MGRS
WMa

WMa
MM
of these monoclonal assays to the original polyclonal FLC
assays. Preliminary studies suggest that they may be
67% in AL, 80% in AH/
Serum EP/immunofixa-

Serum EP/immunofixa-

Serum EP/immunofixa-
tion: 3567% Urine EP/
88% in AH/AHL -Urine

Urine EP/immunofixa-
complementary, although all international guidelines are
AHL -FLC: 76-88% in
Identification of an

AL, 82% in AH/AHL


EP/immunofixation:
tion: 66-80% in AL,

2153% -FLC: 20%

still based on the original FLC assays.


immunofixation:
Table 2 | Main clinical, pathological, and immunological characteristics of glomerular disorders with organized Ig deposits in MGRS

Use of FLC assays in patients with renal impairment


M-protein

tion: 76%

tion: UN requires a degree of diagnostic sensitivity in their interpreta-


FLC: UN tion by either the reporting laboratory or the clinician. This is
a consequence of the close relationship of serum concentra-
tions of FLCs to underlying renal function.52 As renal
Uncommon (peripheral

skin, peripheral nerve,


Extra-renal involve-

impairment progresses, there is a polyclonal elevation in both


peripheral nerve

kappa and lambda FLCs. In addition, in patients with normal


renal function, the physiologically greater production of
nerve, skin)
heart, liver,
Frequent:

Frequent:

polyclonal kappa LC is masked by the more rapid clearance


joints
ment

of monomeric (kappa) LC as compared with larger dimeric


(lambda) LC. In case of renal impairment, there is a subtle
Microtubules 10 to

shift in the FLC ratio. Adoption of the renal-range for the


Parallely arranged

intracellular crys-
tals (crystal-cryo-
branched fibrils

FLC ratio of 0.373.17 (normal range in patients with


1060 nm, with
Ultrastructural

globulinemia)
Randomly ar-

microtubules

hollow core

preserved renal function: 0.261.65) has been shown to


ranged un-

714 nm in
diameter
findings

Extra

increase the reliability of the assays in patients with kidney


90 nm

glomerulonephritis; NS, nephrotic syndrome; Pred., predominantly; UN, unknown; WM, Waldenstroms macroglobulinemia.

impairment.53 Importantly, the delta between the involved


and uninvolved LC is often useful at diagnosis and should be
AH: HC deposits (g1, or g4,

(IgG14IgG24IgG3) (k4l)

Monotypic IgG, IgM, or IgA


endothelial), vascular walls
Granular/smudgy deposits

monitored regularly as it is likely to be the best indicator of


AHL: LC and HC deposits,

Granular deposits in me-


sangium, CW (pred. sub-
or a), with first constant

Monotypic IgG deposits


AL: LC deposits, mostly

in mesangium and CW
domain (CH1) deletion

response to treatment, as validated in AL amyloidosis54 and


- C3, C4, C1q deposits
mostly g l or a k

C3, C4, C1q deposits


IF findings (Ig type)

(pred. subepithelial)

Glomerular thrombi

suggested in most MGRS.12


lambda

RENAL PATHOLOGY
(k4l)

The pathologic diagnosis of renal lesions in MGRS requires


integration of morphologic alterations seen on light micro-
Vascular and tubulo interstitial
and CW deposits (dichroism

Intrarenal vasculitis occasional


Congo-red-positive mesangial

birefringence under polarized

Glomerular thrombi common

scopy (LM), IF, and EM, as well as correlation with the


Mesangial GN with membra-

Interstitial tumoral infiltrate


Light microscopic findings

clinical parameters. IF should be performed using a panel of


involvement common

antibodies specific for the different LC and Ig isotypes. In


glomerular disorders related to deposition of an entire
Endocapillary GN
common (CLL)

monoclonal IgG (immunotactoid glomerulopathy, type I


nous features

cryoglobulinemic glomerulonephritis, PGNMID), or of a


truncated monoclonal heavy chain (heavy chain deposition
MPGN

MPGN
light)

disease (HCDD), AH amyloidosis), IF studies with antibodies


In patients with IgM monoclonal gammopathy.

against IgG subclasses are useful to clearly assess composition


syndrome, AKI, an-
hematuria uncom-
Hypertension and

Possible nephritic

and clonality of deposits. IF and EM are essential to


Renal symptoms

Microhematuria

Microhematuria
Proteinuria, NS

Proteinuria, NS

Proteinuria, NS

determine the specific MGRS lesion, which is crucial for


Hypertension

Hypertension

assessing prognosis and to initiate proper therapy. MGRS


lesions can be divided into two large categories on the basis
mon
CKD

CKD

CKD

uria

of their ultrastructural appearance: those with organized


deposits and those with mostly granular deposits (i.e.,
Type I cryoglobuline-
Glomerular disease

without substructure)55 (Tables 13, Figure 2). In difficult


AHL amyloidosis
AH amyloidosis
AL amyloidosis

cases, sensitive techniques such as immunoEM and laser


ITGN/GOMMID

microdissection/mass spectrometry analyses may be required


to confirm the composition of renal deposits and their
mic

localization.56,57
GN

Kidney International 5
6
Table 3 | Main clinical, pathological, and immunological characteristics of glomerular disorders with non-organized Ig deposits in MGRS
Glomerular Light microscopic Ultrastructural Extrarenal Identification of Hematological and immuno-
disease Renal symptoms pattern IF findings (Ig type) findings involvement an M-protein logical characteristics
review

MIDD Proteinuria, NS Nodular glomerulosclero- Linear deposits along TBM, Amorphous deposits Common, often Serum EP/immuno- MGRS
CKD sis (constant in HCDD) GBM and around arteriolar/ in TBM, GBM, mesan- asymptomatic: fixation: 2576% in Symptomatic MM
Microhematuria Thickened TBM and arterial myocytes gium and arteriolar/ heart, liver, lung LCDD, 80100% in WMa
Hypertension vascular walls LCDD: mostly kappa (Vk4) arterial walls LHCDD, 67100% in Hypocomp. common in g1
HCDD: truncated HC (g1, or HCDD and g3 HCDD
g3, or g4, or a), with CH1 Urine EP/immuno-
deletion. fixation: 4290% in
C3 deposits in g1 and g3 LCDD, 80100% in
HCDD LHCDD, 50100% in
LHCDD: LC truncated HC HCDD
deposits FLC: 100% in LCDD,
LHCDD, HCDD
PGNMID Proteinuria, NS MPGN Granular deposits in mesan- Non-organized gran- None Serum EP/immuno- Usually none
CKD Endocapillary GN gium, CW ular deposits in me- fixation: 30% MGRS
Microhematuria Membranous GN Monotypic IgG deposits: sangium, suben- Urine EP/immuno- MM, B-cell lymphoma, WM:
Hypertension Mesangial GN IgG3 most common, or IgG1, dothelial and/or fixation: 11% rare
or IgG2 (k4l) subepithelial zone FLC: UN Hypocomp. B30%
Rarley, monotypic IgM, IgA,
or LC deposits
C3 C1q deposits
C3 glomerulopathy Proteinuria, NS MPGN Granular C3 deposits in Sausage shaped in- None Serum EP/immuno- MGRS
with monoclonal CKD Mesangial GN mesangium and CW tramembranous and fixation: 100% MM
gammopathy Microhematuria Endocapillary No or paucity of Ig deposits large rounded me- Urine EP: 100% Hypocomp. common, with
Hypertension proliferative GN sangial electron FLC: 75100% low C3 and occasionally anti-
dense deposits in complement factor H auto-
DDD antibody
Ill-defined mesangial,
intramembranous
and subendothelial
electron dense de-
posits in C3GN
Humps common in
DDD and C3GN
Abbreviations: CKD, chronic kidney disease; CW, glomerular capillary walls; DDD, dense deposit disease; EP, electrophoresis; FLC, serum-free light chain assay; GBM, glomerular basement membrane; GN, glomerulonephritis; C3GN,
C3 glomerulonephritis; HCDD, heavy chain deposition disease; Hypocomp., hypocomplementemia; IF, immunofluorescence; Ig, immunoglobulin; LC, immunoglobulin light chains; LCDD, light chain deposition disease; LHCDD,
light and heavy chain deposition disease; MGRS, monoclonal gammopathy of renal significance; MIDD, monoclonal immunoglobulin deposition disease (Randall-type); MM, multiple myeloma; MPGN, membranoproliferative
glomerulonephritis; NS, nephrotic syndrome; PGNMID, proliferative glomerulonephritis with monoclonal immunoglobulin deposits; TBM, tubular basement membrane; UN, unknown; WM, Waldenstroms macroglobulinemia.
a
In patients with IgM monoclonal gammopathy.

Kidney International
F Bridoux et al.: MGRS
F Bridoux et al.: MGRS review

Kidney biopsy a c

Ig or light chain restriction


on immunofluorescence

+ C3 predominant
No further
deposits hematologic
workup
Serum and urine +
monoclonal studies (protein
electrophoresis and Monoclonal
immunofixation, FLC) gammopathy b d
+
Bone marrow C3nef
aspirate and biopsy and anti-H
autoantibodies

Lymph node biopsy* *If bone marrow is negative and


high suspicion for lymphoma

Figure 1 | Proposed algorithm for hematologic workup in


patients with MGRS. MGRS, monoclonal gammopathy of renal
significance; FLC, serum-free light chain assay. Figure 3 | Pathology of renal Igrelated amyloidosis. (a) There is
global marked mesangial and segmental glomerular capillary wall
deposition of acellular non-argyrophilic amyloid deposits (silver stain,
X200). (b) By definition, amyloid deposits should be Congo-red
positive. The figure shows extensive glomerular and vascular and
focal interstitial Congo-red positive amyloid deposits, which produce
MGRS-associated renal lesions anomalous colors (yellow/orange/green) when viewed under
polarized light (X200). (c) On high magnification at the ultrastructural
level, amyloid fibrils appear haphazardly oriented and measure
Organized deposits Non-organized
or inclusions deposits/inclusions
between 7 and 14 nm in diameter (electron microscopy, X46,000).
(d) On immunofluorescence, amyloid deposits appear smudgy and
show light chain restriction. The figure shows global mesangial and
Fibrils Microtubules Crystals or Monoclonal segmental glomerular capillary wall staining for lambda (X400).
inclusions immunoglobulin
deposition disease Staining for kappa was negative (not shown). Ig, immunoglobulin.
(LCDD, LHCDD,
LIght chain proximal HCDD)
Ig-related
Immunotactoid tubulopathy (with or
amyloidosis
(AL, AHL, AH)
GN/GOMMID without Fanconi amyloid is Congo-red positive and produces anomalous
syndrome) Proliferative
GN with mono- colors (yellow/orange/green) under polarized light.60 Small
clonal Ig deposits
Type 1
Crystal-storing (PGNMID) deposits of amyloid may be inconspicuous in hematoxylin
Fibrillary GN cryoglobulinemic
histiocytosis
GN and eosin, PAS, trichrome, and silver stains; hence, a Congo-
C3 glomerulopathy
associated with
red stain should be performed, not only to confirm the
monoclonal
gammopathy
presence of amyloid, but also to rule it out.59 The screening
of Congo-red stained slides under fluorescent light will also
increase the sensitivity of detection of amyloid.59 Renal AIg
Figure 2 | Diagram of MGRS-associated renal lesions.
affects glomeruli and vessels in the vast majority of cases and
AH, immunoglobulin heavy chain amyloidosis; AHL, immunoglobulin
heavy and light chain amyloidosis; AL, immunoglobulin light chain the interstitium in roughly half of the cases, but vascular-
amyloidosis; GN, glomerulonephritis; GOMMID, glomerulonephritis limited AL may rarely occur.40 Ultrastructurally, amyloid
with organized microtubular monoclonal immunoglobulin deposits; deposits appear as randomly arranged, nonbranching fibrils,
HCDD, heavy chain deposition disease; LCDD, light chain deposition
disease; LHCDD, light and heavy chain deposition disease; MGRS,
ranging in thickness between 7 and 14 nm, which can be seen
monoclonal gammopathy of renal significance; PGNMID, proliferative within the mesangium, glomerular basement membranes,
glomerulonephritis with monoclonal immunoglobulin G deposits. vessels, interstitium, and/or tubular basement membranes. A
departure from the haphazard orientation of fibrils is the
formation of amyloid spicules, which result from parallel
MGRS lesions with fibrillar deposits alignment of fibrils in the subepithelial zone perpendicular to
Igrelated amyloidosis. (Figure 3): Igrelated amyloidosis the glomerular basement membrane. The latter is more
(AIg) accounts for 4 80% of cases of renal amyloidosis in common in AIg than other forms of renal amyloidosis.35,61
the US.35 AIg in most cases is derived from fragments of On IF, amyloid deposits appear smudgy with fuzzy borders.
monoclonal AL and rarely from fragments of Ig heavy chains The IF finding of an intense staining for a single AL with
and light chains (AHL) or Ig heavy chains only (AH).58,59 negativity for Ig heavy chains is diagnostic of AL, with
Amyloid has a characteristic appearance on LM: acellular roughly three quarters of renal AL cases expressing lambda
deposits that stain pale eosinophilic on hematoxylin and LC. The diagnosis of AH is based on the presence of intense
eosin, periodic acidSchiff (PAS)-negative or weak, staining for a single Ig heavy chain (most commonly gamma)
trichrome-blue or gray, and silver-negative. By definition with negativity for kappa and lambda LCs, whereas the
and in contrast to the deposits of all other MGRS lesions, diagnosis of AHL is reserved for intense staining for a single

Kidney International 7
review F Bridoux et al.: MGRS

Ig heavy chain and a single AL (most commonly IgGl).58 In a c


early renal AL, the deposits can be inconspicuous by LM and
easily missed by EM but unequivocally identified by IF. IF has
a sensitivity of 82% and a specificity of 100% for diagnosing
AL but has lower sensitivity and specificity for diagnosing Ig
AHL and AH, and therefore a negative or equivocal IF
staining for Ig heavy and/or light chain does not exclude
AIg.35,62 Amyloid typing by laser microdissection followed by b
mass spectrometry has a higher sensitivity and specificity
compared with IF and immunoperoxidase and should be
performed when there is no frozen tissue available for IF,
negative staining for kappa and lambda, equal staining for
kappa and lambda, or bright staining for IgG, IgA, or
IgM.35,63 Laser microdissection/mass spectrometry may also
assist in the distinction between AIg and fibrillary
glomerulonephritis (FGN).64 ImmunoEM may be also Figure 4 | Pathology of immunotactoid glomerulopathy. (a) The
glomerulus shows global glomerular capillary wall thickening and
helpful to assess the composition of amyloid fibrils to mesangial expansion by red-staining immune deposits (trichrome
highlight the association of amyloid fibrils with monoclonal stain, X400). (b) In this case of immunotactoid glomerulopathy with a
Ig deposition and thereby eliminate nonspecific binding of Ig predominantly membranous pattern of injury, there is coarsely
fragments.65 granular global glomerular capillary wall and segmental mesangial
staining for IgG (X400). Similar glomerular staining for lambda was
Non-amyloid FGN. The majority of cases of non-amyloid seen, with negative staining for kappa (not shown). (c) Electron
FGN show polyclonal glomerular Ig deposits, staining microscopy in a different case exhibits large subendothelial and
for IgG (predominantly IgG4 and IgG1), kappa, and lambda mesangial deposits composed of large microtubules (mean 52 nm)
on IF and are not associated with a circulating serum with hollow centers, which are organized in parallel arrays (X9700).
paraprotein.15,66,67 These cases should not be considered a
manifestation of MGRS. However, a minority of cases are
associated with monotypic glomerular deposits and should
be considered a manifestation of MGRS. In a recent zone.15,67,68 On IF, the glomerular deposits generally but
series of 66 cases of FGN, 7 (11%) stained for IgG and a not always stain for IgG (most commonly IgG1) and C3 and
single LC on IF and 3 (5%) stained for IgG without LCs. exhibit LC restriction.
Serum IF performed in 9 of the above 10 cases detected Type I cryoglobulinemic glomerulonephritis. (Figure 5):
a MIg in 8 (89%).66 LM in FGN typically shows Glomerular involvement occurs in 30% of patients with
mesangial hypercellularity, duplication of the glomerular type I cryoglobulinemia.69 On LM, type I cryoglobulinemic
basement membranes, and glomerular non-congophilic glomerulonephritis shows membranoproliferative or endo-
deposits. Ultrastructurally, the deposits are composed capillary proliferative glomerulonephritis with numerous
of randomly oriented fibrils ranging from 9 to 26 nm in intracapillary infiltrating monocytes and large PAS-positive
diameter. In contrast to amyloid fibrils, they rarely form intraluminal immune deposits protein (hyaline)
spicules or affect vessels but can involve tubular basement thrombi.69,70 On EM, the deposits are seen mainly in the
membranes. subendothelial zone and intracapillary lumen and in over half
of cases appear organized (with fibrillar, microtubular, or
MGRS lesions with microtubular substructure fingerprint substructure).70 The organized substructure is
Immunotactoid glomerulopathy. (Figure 4): Immunotac- more commonly seen in the intraluminal deposits. On IF, the
toid glomerulopathy, also referred to as glomerulonephritis glomerular deposits are composed of monoclonal light and
with organized microtubular monoclonal Ig deposits, is heavy chains (most commonly IgG kappa) and comple-
defined by glomerular deposition of microtubules that have ment.71 IF is useful to determine the type of cryoglobulin-
distinct hollow centers at magnification of o30,000, ar- emia present: the glomerular deposits in type II cryoglobulin-
ranged at least focally in parallel arrays, and stain for Igs by emic glomerulonephritis typically exhibit brighter staining
IF, in the absence of clinicopathologic diagnosis of cryoglo- for IgM than IgG and for kappa than lambda (consistent with
bulinemic glomerulonephritis or lupus nephritis.15,67,68 The monoclonal IgM kappa against polyclonal IgG), whereas they
microtubules are usually thicker compared with the fibrils of stain similarly for IgG, IgM, kappa, and lambda in type III
amyloid and FGN, ranging in size from 10 to 90 nm. The two cryoglobulinemic glomerulonephritis (consistent with
most common morphologic patterns of glomerular injury on polyclonal IgGs and IgMs).
LM and EM are membranous glomerulonephritis in which In crystalcryoglobulinemia, monoclonal Ig deposits are
the deposits are primarily present in the subepithelial space highly organized into intracellular and/or extracellular
and membranoproliferative glomerulonephritis in which the crystals in the cytoplasm of glomerular endothelial, mesan-
deposits are preferentially located in the subendothelial gial cells, and/or in glomerular subendothelial spaces and

8 Kidney International
F Bridoux et al.: MGRS review

a b a b

c d c d

200 nm

Figure 5 | Pathology of type I cryoglobulinemic Figure 6 | Pathology of light-chain proximal tubulopathy.


glomerulonephritis. Kidney biopsy from a 65-year-old male (a) Large rod-shaped hypereosinophilic crystals are seen within the
patient with IgA lambda type I cryoglobulinemia. (a) Global cytoplasm of some proximal tubular cells. (H&E, X600). (b) By
endocapillary hypercellularity with numerous hyaline thrombi in immunofluorescence performed on pronase-digested, paraffin-
glomerular capillary lumens (green light trichrome stain, X312). (b) embedded tissue, proximal tubular crystals stain strongly for kappa
The toluidine blue-stained semithin sections revealed crystalline (as shown) with negative lambda (not shown). The intracellular
inclusions within glomerular capillary lumens (X312, and insert crystals failed to stain for kappa or lambda on standard
X1000). (c) By electron microscopy (X2500), subendothelial deposits immunofluorescence on frozen tissue (not shown). (c)
and hyaline thrombi were highly organized with a remarkable grid- Ultrastructurally, the proximal tubular cells are loaded with electron
like appearance on higher magnification (insert, X100,000). (d) dense light-chain crystals with rod, rhomboid, or rectangular shapes.
Immunoelectron microscopy, anti-human lambda light-chain gold The crystals are present predominantly free within the cytoplasm (not
conjugate (X60,000). Numerous gold particles specifically decorated membrane-bound). The proximal tubular brush borders appear intact
crystalline glomerular thrombi. Similar staining was observed with (X1850). (d) In this case of light-chain proximal tubulopathy, the
the anti-alpha conjugate, whereas no significant staining was seen proximal tubular cells are filled with large election dense
with anti-gamma, anti-m, and anti-kappa conjugates (not shown). phagolysosomes, without crystals (electron microscopy, X9700). H&E,
hematoxylin and eosin.

within vascular lumens that may display a typical grid-like eosin and electron dense with occasional mottled
appearance.55 appearance on EM.77 LC proximal tubulopathy without
crystals can be of kappa or lambda types and is not usually
MGRS lesions with deposition of crystals associated with FS.77 A not uncommon diagnostic dilemma is
Light-chain proximal tubulopathy. (Figure 6): LC proximal distinguishing LC proximal tubulopathy without crystals
tubulopathy, previously referred to as the LC Fanconi from the physiologic intracellular proximal tubular
syndrome is characterized by the presence of rod- or trafficking of LC (without pathologic significance). We
rhomboid-shaped hypereosinophilic and PAS-negative crys- recommend that a diagnosis of LC proximal tubulopathy
tals within proximal tubular cells. Ultrastructurally, the without crystals should be reserved to cases associated with
crystals may appear granular or show lattice-like substruc- very large dysmorphic lysosomes, histologic evidence of acute
ture.72 Because of their intracellular localization and and/or chronic proximal tubular injury, marked swelling of
extensive crystallization, standard IF on frozen tissue may proximal tubular cells due to lysosomal indigestion, renal
fail to confirm their LC composition. IF on pronase-digested, insufficiency (not explained by other pathology), tubular
paraffin-embedded tissue, which has an antigen retrieval step, proteinuria (as opposed to pure overflow proteinuria), and/
and immunoEM are much more sensitive techniques, but the or clinical features of partial or complete FS.
latter is not widely used in renal pathology practice.73,74 Crystal-storing histiocytosis. (Figure 7): This is a rare
Patients with LC proximal tubulopathy with crystals may or complication of MM or MGRS, which typically involves the
may not have a complete or partial FS.38,39,75,76 The bone marrow but can also affect several extramedullary sites,
pathologic LC in this lesion is almost always kappa and including the kidney, peri-renal fat, lungs, and cornea.
restricted to the Vk1 subgroup. A more recently described Kidney biopsy reveals intracytoplasmic eosinophilic LC
pathologic variant of light-chain proximal tubulopathy called crystalline inclusions within interstitial histiocytes and
LC proximal tubulopathy without crystals manifests LC- occasionally within proximal tubular cells and podocytes.79
containing phagolysosomes within proximal tubular cells As in LC proximal tubulopathy with crystals, standard IF on
without crystal formation.72,77,78 The non-crystalline frozen tissue sometimes fails to demonstrate their LC
inclusions appear hypereosinophilic on hematoxylin and composition, which requires IF on pronase-digested

Kidney International 9
review F Bridoux et al.: MGRS

subtype, in which the deposits contain monoclonal LC only


(kappa in most cases), light and heavy chain deposition
disease (LHCDD), in which the deposits are composed of
monoclonal light and heavy chains (most commonly IgG
kappa), and HCDD, in which the deposits are derived from
monoclonal heavy chains only (most frequently gamma),
which always lack the first constant (CH1) domain. LM
shows thickening of tubular basement membranes in most
cases and nodular mesangial sclerosis in 2/3 of cases,
accompanied by variable degrees of tubular atrophy, inter-
stitial fibrosis, and inflammation.7,9,81 It has a very distinctive
appearance on IF: diffuse linear staining along basement
membranes of glomeruli, tubules, and vascular myocytes for
Figure 7 | Pathology of crystal-storing histiocytosis. The figure kappa or lambda only in LCDD, for a single Ig heavy chain
shows numerous light-chain crystals with rod, needle, or rhomboid
shapes, within the cytoplasm of interstitial infiltrating histiocytes. The with LC restriction in LHCDD, or for a single class of Ig with
tubule depicted in the upper portion of the image does not exhibit CH1 domain deletion and no corresponding LC in HCDD.
intracytoplasmic or intraluminal crystals (electron microscopy, The IF deposits correspond to punctate powdery electron-
X1850).
dense deposits along the inner aspect of glomerular basement
membranes and outer aspect of tubular basement
membranes on EM. Glomerular crescents may be seen,
particularly in alpha HCDD.82
a b Proliferative glomerulonephritis with monoclonal IgG depos-
its. (Figure 9): LM in PGNMID generally shows membra-
noproliferative or endocapillary proliferative glomerulo-
nephritis, with or without membranous features, and less
commonly pure mesangial proliferative glomerulo-
nephritis.27,83 EM exhibits granular, non-organized
deposits, typically in a subendothelial and mesangial
distribution, with or without subepithelial deposits. By
c d
definition, and in contrast to immune complex type
glomerulonephritis, the glomerular deposits on IF appear
monoclonal, staining for a single LC isotype and a single IgG
heavy chain subtype (most commonly IgG3).27 PGNMID
patients with monoclonal IgG3 deposits are less likely
to have a detectable circulating paraprotein compared
Figure 8 | Pathology of renal monoclonal immunoglobulin with those with monoclonal IgG1 or IgG2 deposits.27
deposition disease. (a) The glomerulus shows nodular mesangial Contrary to Randall-type HCDD and LHCDD, the deposits
sclerosis, which is seen in most but not all cases of MIDD. The nodules in PGNMID are restricted to glomeruli (without
are PAS-positive similar to the nodules of diabetic glomerulosclerosis
(PAS, X400). (b) The diagnostic ultrastructural finding in MIDD is finely extraglomerular or extrarenal involvement) and contain an
granular electron dense deposits involving the outer aspect of the intact MIg. Of note, there have been rare reported cases
tubular basement membranes (as seen in this figure) and the inner of glomerulonephritis in which the glomerular deposits are
aspect of the glomerular basement membranes (not shown) (electron
microscopy, X2400). (c and d) In this case of LCDD kappa type,
composed of MIg components other than IgG, including
immunofluorescence reveals diffuse linear glomerular and tubular IgM, IgA, or LCs only.46,8486
basement membranes staining for kappa (c) with negative staining C3 glomerulopathy with monoclonal gammopathy. (Fig-
for lambda (d; X100 for c and d). LCDD, light chain deposition ure 10): C3 glomerulopathy encompasses two pathologic
disease; MIDD, monoclonal immunoglobulin deposition disease. PAS,
periodic acidSchiff.
entities, C3 glomerulonephritis and dense deposit disease,
both of which are due to dysregulation of the complement
alternative pathway (CAP; e.g., mutations or functional inhi-
paraffin tissue or evaluation by the immunoperoxidase bition of complement-regulating proteins) and are charac-
method. Crystal-storing histiocytosis may coexist with LC terized by glomerular C3 deposition with absent or scanty Ig
proximal tubulopathy and/or cast nephropathy.80 deposition.87 LM in C3 glomerulopathy shows mesangial
proliferative, membranoproliferative, or endocapillary pro-
MGRS lesions with granular (non-organized) deposits liferative glomerulonephritis. Dense deposit disease is
MIg deposition disease of the Randall type (MIDD). distinguished from C3 glomerulonephritis on the ultrastruc-
(Figure 8): MIDD includes three subtypes, depending on tural level: the former shows highly electron dense sausage-
the composition of deposits: LCDD, the most common like deposits that segmentally infiltrate the glomerular

10 Kidney International
F Bridoux et al.: MGRS review

2 m

lgG3 Kappa Lambda

Figure 9 | Pathology of proliferative glomerulonephritis with monoclonal IgG deposits. (a) Glomeruli exhibit marked global mesangial and
segmental endocapillary hypercellularity (H&E, X200). (b) On electron microscopy, there are large mesangial, intramembranous, and
subepithelial deposits, together with segmental duplication of the glomerular basement membrane. The electron dense deposits appear
granular (without substructure; X6000). (ce) Glomeruli in this case of PGNMID show bright global mesangial and glomerular capillary wall
staining for IgG3 (c) and kappa (d). Glomeruli are negative for lambda (e), IgA, IgM, IgG1, IgG2, and IgG4 (not shown). Note the lack of tubular
basement membrane deposits, which is quite different from monoclonal immunoglobulin deposition disease of the Randall type (X100 for ce).
H&E, hematoxylin and eosin; Ig, immunoglobulin; PGNMID, proliferative glomerulonephritis with monoclonal immunoglobulin G deposits.

Figure 10 | Pathology of C3 glomerulopathy. ac are from a 69-year-old patient with IgG lambda MGRS and C3 glomerulonephritis.
Complement pathway workup showed CFH risk polymorphism (Y402H). On light microscopy, the glomerulus shows marked global mesangial
and segmental endocapillary hypercellularity with intracapillary infiltrating lymphocytes, monocytes, and some neutrophils (a; PAS, X200). On
immunofluorescence, there is global granular mesangial and glomerular capillary loop staining for C3 (b, X200). Glomeruli were negative for
IgG, IgA, IgM, kappa, and lambda in this case (not shown). Ultrastructurally, there are large electron dense mesangial deposits and scattered
subepithelial deposits (arrows; c, X7830). df are from a 59-year-old male with IgG kappa MGRS and dense deposit disease. Complement
pathway workup was positive for FH autoantibody. The glomerulus depicted shows global mesangial hypercellularity and segmental occlusion
of peripheral capillaries by endocapillary hypercellularity and influx of inflammatory cells (arrows). The glomerular basement membranes
appear segmentally thickened (d; H&E, X200). Immunofluorescence highlights global granular to semilinear glomerular capillary wall and
mesangial C3 deposits, with linear staining of Bowmans capsule (e; X200). The defining feature of dense deposit disease is sausage-like
thickening of the glomerular basement membranes by highly electron dense intramembranous deposits. Large rounded mesangial electron
dense deposits are also seen (f; X6000). CFH, complement factor H; H&E, hematoxylin and eosin; MGRS, monoclonal gammopathy of renal
significance; PAS, periodic acidSchiff.

Kidney International 11
review F Bridoux et al.: MGRS

basement membrane lamina densa, whereas the latter shows DISCLOSURE


less electron dense, ill-defined granular deposits in the All the authors declared no competing interests.
subendothelial space, subepithelial space, and/or
mesangium. A subset of patients with dense deposit disease REFERENCES
or C3 glomerulonephritis have a demonstrable circulating 1. Solomon A, Weiss DT, Kattine AA. Nephrotoxic potential of Bence Jones
proteins. N Engl J Med 1991; 324: 18451851.
MIg in the blood and evidence of a plasma cell dyscrasia in
2. Alexanian R, Barlogie B, Dixon D. Renal failure in multiple myeloma.
the bone marrow.8890 In contrast to all other MGRS lesions, Pathogenesis and prognostic implications. Arch Intern Med 1990; 150:
the MIg in C3 glomerulopathy is not deposited in the kidney. 16931695.
3. Drayson M, Begum G, Basu S et al. Effects of paraprotein heavy and light
In these patients, the MIg likely causes glomerulonephritis chain types and free light chain load on survival in myeloma: an analysis
indirectly through continuous CAP activation, such as by of patients receiving conventional-dose chemotherapy in Medical
inhibiting the complement-regulating proteins.24,89,91 Several Research Council UK multiple myeloma trials. Blood 2006; 108:
20132019.
arguments strongly suggest a causal relationship between 4. Morel-Maroger L, Basch A, Danon F et al. Pathology of the kidney in
monoclonal gammopathy and C3 glomerulopathy. First, Waldenstroms macroglobulinemia. Study of sixteen cases. N Engl J Med
3050% of patients 450 years of age with C3 glomerulopathy 1970; 283: 123129.
5. Merlini G, Stone MJ. Dangerous small B-cell clones. Blood 2006; 108:
have a detectable monoclonal Ig,89 a frequency that largely 25202530.
exceeds that of MGUS in the general population (3.2% in 6. Kourelis TV, Kumar SK, Gertz MA et al. Coexistent multiple myeloma or
increased bone marrow plasma cells define equally high-risk populations
patients aged over 50 years).92 Second, as mentioned above, in patients with immunoglobulin light chain amyloidosis. J Clin Oncol
monoclonal Igs may induce CAP through autoantibody 2013; 31: 43194324.
activity against factor H. In a patient with MPGN Meri24 and 7. Nasr SH, Valeri AM, Cornell LD et al. Renal monoclonal immunoglobulin
deposition disease: a report of 64 patients from a single institution. Clin J
Jokiranta25 isolated and characterized a dimeric monoclonal Am Soc Nephrol 2012; 7: 231239.
Vl3 LC, which behaved as a mini autoantibody to factor H. 8. Kyle RA, Durie BG, Rajkumar SV et al. Monoclonal gammopathy of
By binding to the short consensus repeat domain 3 at the undetermined significance (MGUS) and smoldering (asymptomatic)
multiple myeloma: IMWG consensus perspectives risk factors for
N-terminal end of factor H, this dimer blocked the progression and guidelines for monitoring and management. Leukemia
interaction between factor H and C3b, thereby inhibiting 2010; 24: 11211127.
9. Pozzi C, DAmico M, Fogazzi GB et al. Light chain deposition disease with
the activity of factor H and inducing uncontrolled CAP renal involvement: clinical characteristics and prognostic factors. Am J
activation.24,25 Third, disappearance of anti-factor H auto- Kidney Dis 2003; 42: 11541163.
antibodies with hematological response after chemotherapy 10. Leung N, Bridoux F, Hutchison CA et al. Monoclonal gammopathy of renal
significance: when MGUS is no longer undetermined or insignificant.
has been described in some patients with C3 Blood 2012; 120: 42924295.
glomerulopathy.90 However, the role of MIg in CAP 11. Jaccard A, Moreau P, Leblond V et al. High-dose melphalan versus
activation in C3 glomerulopathy remains to be formally melphalan plus dexamethasone for AL amyloidosis. N Engl J Med 2007;
357: 10831093.
demonstrated by the few reference laboratories capable of 12. Fermand JP, Bridoux F, Kyle RA et al. How I treat monoclonal
performing appropriate functional assays. gammopathy of renal significance (MGRS). Blood 2013; 122:
35833590.
13. Leung N, Dispenzieri A, Fervenza FC et al. Renal response after high-dose
SUMMARY melphalan and stem cell transplantation is a favorable marker in patients
The pathologic spectrum of renal diseases in MGRS is wide with primary systemic amyloidosis. Am J Kidney Dis 2005; 46: 270277.
14. Lorenz EC, Gertz MA, Fervenza FC et al. Long-term outcome of
and kidney biopsy is required to determine the exact nature autologous stem cell transplantation in light chain deposition disease.
of the lesion and to evaluate the severity of renal disease. The Nephrol Dial Transplant 2008; 23: 20522057.
monoclonal protein may cause kidney damage directly 15. Bridoux F, Hugue V, Coldefy O et al. Fibrillary glomerulonephritis and
immunotactoid (microtubular) glomerulopathy are associated with
through deposition or precipitation, as in AL amyloidosis, distinct immunologic features. Kidney Int 2002; 62: 17641775.
MIDD, and light-chain proximal tubulopathy, or indirectly 16. Guiard E, Karras A, Plaisier E et al. Patterns of noncryoglobulinemic
through an autoantibody activity, as in C3 glomerulopathy glomerulonephritis with monoclonal Ig deposits: correlation with IgG
subclass and response to rituximab. Clin J Am Soc Nephrol 2011; 6:
with monoclonal gammopathy. Because of the small amount 16091616.
and/or rapid deposition in the kidney, the paraprotein may 17. Pinney JH, Lachmann HJ, Bansi L et al. Outcome in renal Al amyloidosis
after chemotherapy. J Clin Oncol 2011; 29: 674681.
be undetectable in the blood by serum EP/immunofixation 18. Czarnecki PG, Lager DJ, Leung N et al. Long-term outcome of kidney
or FLC and therefore may require more sensitive techniques. transplantation in patients with fibrillary glomerulonephritis or
Early recognition of MGRS is crucial, as it strongly impacts monoclonal gammopathy with fibrillary deposits. Kidney Int 2009; 75:
420427.
renal prognosis. A common problem in clinical nephro- 19. Leung N, Lager DJ, Gertz MA et al. Long-term outcome of renal
logy practice is to convince hematologists to initiate swift transplantation in light-chain deposition disease. Am J Kidney Dis 2004;
and efficient chemotherapy for patients with MGRS, 43: 147153.
20. Nasr SH, Sethi S, Cornell LD et al. Proliferative glomerulonephritis with
who by definition do not fulfill the criteria for MM. Hence, monoclonal IgG deposits recurs in the allograft. Clin J Am Soc Nephrol
there is a need to increase clinicians awareness of MGRS, 2011; 6: 122132.
21. Pinney JH, Lachmann HJ, Sattianayagam PT et al. Renal transplantation in
as more widespread acceptance of this concept should systemic amyloidosis-importance of amyloid fibril type and precursor
result in less delay in appropriate therapy. A consensus on protein abundance. Am J Transplant 2013; 13: 433441.
the pathological definition of the MGRS spectrum is needed 22. Zand L, Lorenz EC, Cosio FG et al. Clinical Findings, Pathology, and
Outcomes of C3GN after Kidney Transplantation. J Am Soc Nephrol 2013.
in order to design future collaborative studies in these 23. Sanada S, Ookawara S, Karube H et al. Marked recovery of severe renal
disorders. lesions in POEMS syndrome with high-dose melphalan therapy

12 Kidney International
F Bridoux et al.: MGRS review

supported by autologous blood stem cell transplantation. Am J Kidney Dis 49. Campbell JP, Cobbold M, Wang Y et al. Development of a highly-sensitive
2006; 47: 672679. multi-plex assay using monoclonal antibodies for the simultaneous
24. Meri S, Koistinen V, Miettinen A et al. Activation of the alternative measurement of kappa and lambda immunoglobulin free light chains in
pathway of complement by monoclonal lambda light chains in serum and urine. J Immunol Methods 2013; 391: 113.
membranoproliferative glomerulonephritis. J Exp Med 1992; 175: 50. Nakano T, Nagata A. ELISAs for free human immunoglobulin light chains
939950. in serum: improvement of assay specificity by using two specific
25. Jokiranta TS, Solomon A, Pangburn MK et al. Nephritogenic lambda light antibodies in a sandwich detection method. J Immunol Methods 2004;
chain dimer: a unique human miniautoantibody against complement 293: 183189.
factor H. J Immunol 1999; 163: 45904596. 51. te Velthuis H, Knop I, Stam P et al. N Latex FLC-new monoclonal high-
26. Debiec H, Hanoy M, Francois A et al. Recurrent membranous performance assays for the determination of free light chain kappa and
nephropathy in an allograft caused by IgG3kappa targeting the PLA2 lambda. Clin Chem Lab Med 2011; 49: 13231332.
receptor. J Am Soc Nephrol 2012; 23: 19491954. 52. Hutchison CA, Harding S, Hewins P et al. Quantitative assessment of
27. Nasr SH, Satoskar A, Markowitz GS et al. Proliferative glomerulonephritis serum and urinary polyclonal free light chains in patients with chronic
with monoclonal IgG deposits. J Am Soc Nephrol 2009; 20: 20552064. kidney disease. Clin J Am Soc Nephrol 2008; 3: 16841690.
28. Kyle RA, Gertz MA, Greipp PR et al. A trial of three regimens for primary 53. Hutchison CA, Plant T, Drayson M et al. Serum free light chain
amyloidosis: colchicine alone, melphalan and prednisone, and melphalan, measurement aids the diagnosis of myeloma in patients with severe renal
prednisone, and colchicine. N Engl J Med 1997; 336: 12021207. failure. BMC Nephrol 2008; 9: 11.
29. Palladini G, Dispenzieri A, Gertz MA et al. New criteria for response to 54. Palladini G, Dispenzieri A, Gertz MA et al. New criteria for response to
treatment in immunoglobulin light chain amyloidosis based on free light treatment in immunoglobulin light chain amyloidosis based on free light
chain measurement and cardiac biomarkers: impact on survival chain measurement and cardiac biomarkers: impact on survival
outcomes. J Clin Oncol 2012; 30: 45414549. outcomes. J Clin Oncol 2012; 30: 45414549.
30. Venner CP, Lane T, Foard D et al. Cyclophosphamide, bortezomib, and 55. Touchard G. Ultrastructural pattern and classification of renal monoclonal
dexamethasone therapy in AL amyloidosis is associated with high clonal immunoglobulin deposits. In: Touchard G, Aucouturier P, Hermine O,
response rates and prolonged progression-free survival. Blood 2012; 119: Ronco P (eds) Monoclonal gammopathies and the kidney. Kluwer
43874390. Academic Publishers: Dordrecht ; Boston, 2003 pp 95120.
31. Picken MM. Amyloidosis-where are we now and where are we heading? 56. Gu X, Herrera GA. Light-chain-mediated acute tubular interstitial
Arch Pathol Lab Med 2010; 134: 545551. nephritis: a poorly recognized pattern of renal disease in patients with
32. Merlini G, Wechalekar AD, Palladini G. Systemic light chain amyloidosis: plasma cell dyscrasia. Arch Pathol Lab Med 2006; 130: 165169.
an update for treating physicians. Blood 2013; 121: 51245130. 57. Jain D, Green JA, Bastacky S et al. Membranoproliferative
33. Lachmann HJ, Booth DR, Booth SE et al. Misdiagnosis of hereditary glomerulonephritis: the role for laser microdissection and mass
amyloidosis as AL (primary) amyloidosis. N Engl J Med 2002; 346: spectrometry. Am J Kidney Dis 2014; 63: 324328.
17861791. 58. Nasr SH, Said SM, Valeri AM et al. The diagnosis and characteristics of
34. Said SM, Sethi S, Valeri AM et al. Characterization and outcomes of renal renal heavy-chain and heavy/light-chain amyloidosis and their
leukocyte chemotactic factor 2-associated amyloidosis. Kidney Int 2014; comparison with renal light-chain amyloidosis. Kidney Int 2013; 83:
86: 370377. 463470.
35. Said SM, Sethi S, Valeri AM et al. Renal amyloidosis: origin and 59. Picken MM. Modern approaches to the treatment of amyloidosis: the
clinicopathologic correlations of 474 recent cases. Clin J Am Soc Nephrol critical importance of early detection in surgical pathology. Adv Anat
2013; 8: 15151523. Pathol 2013; 20: 424439.
36. Fish R, Pinney J, Jain P et al. The incidence of major hemorrhagic 60. Howie AJ, Brewer DB. Optical properties of amyloid stained by Congo
complications after renal biopsies in patients with monoclonal red: history and mechanisms. Micron 2009; 40: 285301.
gammopathies. Clin J Am Soc Nephrol 2010; 5: 19771980. 61. Shiiki H, Shimokama T, Yoshikawa Y et al. Renal amyloidosis. Correlations
37. Leung N, Gertz M, Kyle RA et al. Urinary albumin excretion patterns of between morphology, chemical types of amyloid protein and clinical
patients with cast nephropathy and other monoclonal gammopathy- features. Virchows Arch A Pathol Anat Histopathol 1988; 412:
related kidney diseases. Clin J Am Soc Nephrol 2012; 7: 19641968. 197204.
38. Bridoux F, Sirac C, Hugue V et al. Fanconis syndrome induced by a 62. Picken MM. Immunoglobulin light and heavy chain amyloidosis AL/AH:
monoclonal Vkappa3 light chain in Waldenstroms macroglobulinemia. renal pathology and differential diagnosis. Contrib Nephrol 2007; 153:
Am J Kidney Dis 2005; 45: 749757. 135155.
39. Messiaen T, Deret S, Mougenot B et al. Adult Fanconi syndrome 63. Sethi S, Vrana JA, Theis JD et al. Laser microdissection and mass
secondary to light chain gammopathy. Clinicopathologic heterogeneity spectrometry-based proteomics aids the diagnosis and typing of renal
and unusual features in 11 patients. Medicine 2000; 79: 135154. amyloidosis. Kidney Int 2012; 82: 226234.
40. Eirin A, Irazabal MV, Gertz MA et al. Clinical features of patients with 64. Sethi S, Theis JD, Vrana JA et al. Laser microdissection and proteomic
immunoglobulin light chain amyloidosis (AL) with vascular-limited analysis of amyloidosis, cryoglobulinemic GN, fibrillary GN, and
deposition in the kidney. Nephrol Dia Transplant 2012; 27: 10971101. immunotactoid glomerulopathy. Clin J Am Soc Nephrol 2013; 8: 915921.
41. Sicard A, Karras A, Goujon JM et al. Light chain deposition disease 65. Herrera GA, Turbat-Herrera EA. Ultrastructural immunolabeling in the
without glomerular proteinuria: a diagnostic challenge for the diagnosis of monoclonal light-and heavy-chain-related renal diseases.
nephrologist. Nephrol Dial Transplant 2014; 29: 18941902. Ultrastruct Pathol 2010; 34: 161173.
42. Katzmann JA, Kyle RA, Benson J et al. Screening panels for detection of 66. Nasr SH, Valeri AM, Cornell LD et al. Fibrillary glomerulonephritis: a report
monoclonal gammopathies. Clin Chem 2009; 55: 15171522. of 66 cases from a single institution. Clin J Am Soc Nephrol 2011; 6:
43. Palladini G, Russo P, Bosoni T et al. Identification of amyloidogenic light 775784.
chains requires the combination of serum-free light chain assay with 67. Rosenstock JL, Markowitz GS, Valeri AM et al. Fibrillary and
immunofixation of serum and urine. Clin Chem 2009; 55: 499504. immunotactoid glomerulonephritis: distinct entities with different clinical
44. Anderson KC, Alsina M, Bensinger W et al. NCCN clinical practice and pathologic features. Kidney Int 2003; 63: 14501461.
guidelines in oncology: multiple myeloma. J Natl Compr Canc Netw 2009; 68. Nasr SH, Fidler ME, Cornell LD et al. Immunotactoid glomerulopathy:
7: 908942. clinicopathologic and proteomic study. Nephrol Dial Transplant 2012; 27:
45. Dispenzieri A, Kyle R, Merlini G et al. International Myeloma Working 41374146.
Group guidelines for serum-free light chain analysis in multiple myeloma 69. Terrier B, Karras A, Kahn JE et al. The spectrum of type I cryoglobulinemia
and related disorders. Leukemia 2009; 23: 215224. vasculitis: new insights based on 64 cases. Medicine (Baltimore) 2013; 92:
46. Sethi S, Zand L, Leung N et al. Membranoproliferative glomerulonephritis 6168.
secondary to monoclonal gammopathy. Clin J Am Soc Nephrol 2010; 5: 70. Nasr SH, Markowitz GS, Reddy BS et al. Dysproteinemia, proteinuria, and
770782. glomerulonephritis. Kidney Int 2006; 69: 772775.
47. Bradwell AR, Carr-Smith HD, Mead GP et al. Highly sensitive, automated 71. Karras A, Noel LH, Droz D et al. Renal involvement in monoclonal (type I)
immunoassay for immunoglobulin free light chains in serum and urine. cryoglobulinemia: two cases associated with IgG3 kappa cryoglobulin.
Clin Chem 2001; 47: 673680. Am J Kidney Dis 2002; 40: 10911096.
48. Davern S, Tang LX, Williams TK et al. Immunodiagnostic capabilities of 72. Kapur U, Barton K, Fresco R et al. Expanding the pathologic spectrum of
anti-free immunoglobulin light chain monoclonal antibodies. Am J Clin immunoglobulin light chain proximal tubulopathy. Arch Pathol Lab Med
Pathol 2008; 130: 702711. 2007; 131: 13681372.

Kidney International 13
review F Bridoux et al.: MGRS

73. Nasr SH, Galgano SJ, Markowitz GS et al. Immunofluorescence on 83. Nasr SH, Markowitz GS, Stokes MB et al. Proliferative glomerulonephritis
pronase-digested paraffin sections: a valuable salvage technique for renal with monoclonal IgG deposits: a distinct entity mimicking immune-
biopsies. Kidney Int 2006; 70: 21482151. complex glomerulonephritis. Kidney Int 2004; 65: 8596.
74. Gu X, Barrios R, Cartwright J et al. Light chain crystal deposition as a 84. Yahata M, Nakaya I, Takahashi S et al. Proliferative glomerulonephritis
manifestation of plasma cell dyscrasias: the role of immunoelectron with monoclonal IgM deposits without Waldenstroms
microscopy. Hum Pathol 2003; 34: 270277. macroglobulinemia: case report and review of the literature. Clin Nephrol
75. Elliott MR, Cortese C, Moreno-Aspitia A et al. Plasma cell dyscrasia causing 2012; 77: 254260.
light chain tubulopathy without Fanconi syndrome. Am J Kidney Dis 2010; 85. Soares SM, Lager DJ, Leung N et al. A proliferative glomerulo-
55: 11361141. nephritis secondary to a monoclonal IgA. Am J Kidney Dis 2006; 47:
76. Said SM, Assaad AM, Cerda J et al. Light chain tubulopathy without 342349.
Fanconi syndrome. Nephrol Dial Transplant 2006; 21: 35893590. 86. Alpers CE, Tu WH, Hopper J Jr. et al. Single light chain subclass (kappa
77. Larsen CP, Bell JM, Harris AA et al. The morphologic spectrum and clinical chain) immunoglobulin deposition in glomerulonephritis. Hum Pathol
significance of light chain proximal tubulopathy with and without crystal 1985; 16: 294304.
formation. Mod Pathol 2011; 24: 14621469. 87. Pickering MC, DAgati VD, Nester CM et al. C3 glomerulopathy: consensus
78. Herrera GA. Renal lesions associated with plasma cell dyscrasias: practical report. Kidney Int 2013; 84: 10791089.
approach to diagnosis, new concepts, and challenges. Arch Pathol Lab 88. Sethi S, Sukov WR, Zhang Y et al. Dense deposit disease associated with
Med 2009; 133: 249267. monoclonal gammopathy of undetermined significance. Am J Kidney Dis
79. El Hamel C, Thierry A, Trouillas P et al. Crystal-storing histiocytosis with 2010; 56: 977982.
renal Fanconi syndrome: pathological and molecular characteristics 89. Zand L, Kattah A, Fervenza FC et al. C3 glomerulonephritis associated
compared with classical myeloma-associated Fanconi syndrome. Nephrol with monoclonal gammopathy: a case series. Am J Kidney Dis 2013; 62:
Dial Transplant 2010; 25: 29822990. 506514.
80. Stokes MB, Aronoff B, Siegel D et al. Dysproteinemia-related nephropathy 90. Bridoux F, Desport E, Fremeaux-Bacchi V et al. Glomerulonephritis with
associated with crystal-storing histiocytosis. Kidney Int 2006; 70: 597602. isolated C3 deposits and monoclonal gammopathy: a fortuitous
81. Lin J, Markowitz GS, Valeri AM et al. Renal monoclonal immunoglobulin association? Clin Am Soc Nephrol 2011; 6: 21652174.
deposition disease: the disease spectrum. J Am Soc Nephrol 2001; 12: 91. Sethi S, Rajkumar SV. Monoclonal gammopathy-associated proliferative
14821492. glomerulonephritis. Mayo Clin Proc 2013; 88: 12841293.
82. Alexander MP, Nasr SH, Watson DC et al. Renal crescentic alpha heavy 92. Kyle RA, Therneau TM, Rajkumar SV et al. Prevalence of monoclonal
chain deposition disease: a report of 3 cases and review of the literature. gammopathy of undetermined significance. N Engl J Med 2006; 354:
Am J Kidney Dis 2011; 58: 621625. 13621369.

14 Kidney International

Das könnte Ihnen auch gefallen