Case Report Nephrotic Syndrome Secondary to Proliferative Glomerulonephritis with Monoclonal Immunoglobulin Deposits of Lambda Light Chain

Similar documents
Interesting case seminar: Native kidneys Case Report:

Monoclonal Gammopathies and the Kidney. Tibor Nádasdy, MD The Ohio State University, Columbus, OH

Case Report A Case of Proliferative Glomerulonephritis with Monoclonal IgG Deposits That Showed Predominantly Membranous Features

Renal Pathology 1: Glomerulus. With many thanks to Elizabeth Angus PhD for EM photographs

Jo Abraham MD Division of Nephrology University of Utah

A Case of IgG2 Heavy Chain Deposition Disease in a Patient with Kappa Positive Plasma Cell Dyscrasia

Surgical Pathology Report

Glomerular pathology in systemic disease

Case 3. ACCME/Disclosure. Laboratory results. Clinical history 4/13/2016

Dr Ian Roberts Oxford. Oxford Pathology Course 2010 for FRCPath Illustration-Cellular Pathology. Oxford Radcliffe NHS Trust

RENAL HISTOPATHOLOGY

Mayo Clinic/ RPS Consensus Report on Classification, Diagnosis, and Reporting of Glomerulonephritis

Ordering Physician. Collected REVISED REPORT. Performed. IgG IF, Renal MCR. Lambda IF, Renal MCR. C1q IF, Renal. MCR Albumin IF, Renal MCR

A clinical syndrome, composed mainly of:

Clinicopathological analysis of proliferative glomerulonephritis with monoclonal IgG deposits in 5 renal allografts

Histopathology: Glomerulonephritis and other renal pathology

CASE 3 AN UNUSUAL CASE OF NEPHROTIC SYNDROME

Glomerular diseases with organized deposits

Journal of Nephropathology

FIBRILLARY GLOMERULONEPHRITIS DIAGNOSTIC CRITERIA, PITFALLS, AND DIFFERENTIAL DIAGNOSIS

Dr Ian Roberts Oxford. Oxford Pathology Course 2010 for FRCPath Illustration-Cellular Pathology. Oxford Radcliffe NHS Trust

Glomerular diseases mostly presenting with Nephritic syndrome

Classification of Glomerular Diseases and Defining Individual Glomerular Lesions: Developing International Consensus

Pathology of Complement Mediated Renal Disease

GOODPASTURE'S SYNDROME WITH CONCOMITANT IMMUNE COMPLEX MIXED MEMBRANOUS AND PROLIFERATIVE GLOMERULONEFRITIS

Glomerular Pathology- 1 Nephrotic Syndrome. Dr. Nisreen Abu Shahin

Overview of glomerular diseases

Tarek ElBaz, MD. Prof. Internal Medicine Chief, Division of Renal Medicine Al Azhar University President, ESNT

Case Presentation Turki Al-Hussain, MD

Monoclonal gammopathies consist of. Monoclonal GammopathyeAssociated Proliferative Glomerulonephritis REVIEW

A case of heavy chain deposition disease complicated by acquired angioedema.

Disorders of the kidney. Urine analysis. Nephrotic and nephritic syndrome.

THE KIDNEY AND SLE LUPUS NEPHRITIS

Proliferative Glomerulonephritis with Monoclonal IgG Deposits Recurs in the Allograft

THE URINARY SYSTEM. The cases we will cover are:

THE URINARY SYSTEM. The cases we will cover are:

Rituximab treatment for fibrillary glomerulonephritis

Renal manifestations of IgG4-related systemic disease

RECURRENT AND DE NOVO RENAL DISEASES IN THE ALLOGRAFT

Glomerular pathology-2 Nephritic syndrome. Dr. Nisreen Abu Shahin

Lab 3, case 1. Is this an example of nephrotic or nephritic syndrome? Why? Which portion of the nephron would you expect to be abnormal?

Focal Segmental Glomerulosclerosis and the Nephro6c Syndrome Dr. A. Gangji Dr. P. Marge>s. Part 1: Clinical

Jon Von Visger 1, Clarissa Cassol 2, Uday Nori 1, Gerardo Franco-Ahumada 1, Tibor Nadasdy 2 and Anjali A. Satoskar 2*

PATTERNS OF RENAL INJURY

RECURRENT AND DE NOVO RENAL DISEASES IN THE ALLOGRAFT. J. H. Helderman,MD,FACP,FAST

Approach to Glomerular Diseases: Clinical Presentation Nephrotic Syndrome Nephritis

Glomerular Diseases. Anna Vinnikova, MD Nephrology

Nephrotic syndrome minimal change disease vs. IgA nephropathy. Hadar Meringer Internal medicine B Sheba

Index. electron microscopy, 81 immunofluorescence microscopy, 80 light microscopy, 80 Amyloidosis clinical setting, 185 etiology/pathogenesis,

Immune profile of IgA-dominant diffuse proliferative glomerulonephritis

substance staining with IgG, C3 and IgA (trace) Linear deposition of IgG(+), IgA.M(trace) and C3(+++) at the DEJ

CHAPTER 2. Primary Glomerulonephritis

Case Presentation Turki Al-Hussain, MD

Elevated Serum Creatinine, a simplified approach

CJASN epress. Published on September 28, 2010 as doi: /CJN

Nephrology Grand Rounds. Mansi Mehta November 24, 2015

Proliferative Glomerulonephritis with Monoclonal IgG Deposits

Hematology 101. Rachid Baz, M.D. 5/16/2014

C3 GLOMERULOPATHIES. Budapest Nephrology School Zoltan Laszik

Familial DDD associated with a gain-of-function mutation in complement C3.

LIGHT CHAIN DISEASE B. DHANALAKSHMI 1 & V. HEMAVATHY 2

Laboratory Examination

A Case of Immunotactoid Glomerulopathy with Rapid Progression to End-Stage Renal Disease

M-Protien, what to do next? Ismail A Sharif MD, FRCPc Internal Medicine Day 22 nd April 2016

Crescentic Glomerulonephritis (RPGN)

RENAL EVENING SPECIALTY CONFERENCE

Glomerulonephritis. Dr Rodney Itaki Anatomical Pathology Discipline.

CHAPTER 2 PRIMARY GLOMERULONEPHRITIS

Year 2004 Paper one: Questions supplied by Megan

C1q nephropathy the Diverse Disease

Multiple Myeloma Advances for clinical pathologists & histopathologists

Long-term follow-up of juvenile acute nonproliferative glomerulitis (JANG)

Expanding Spectrum of Diseases Associated with Plasma Cell Dyscrasias

CASE 4 A RARE CASE OF INTRALUMINAL GLOMERULAR CAPILLARY DEPOSITS

Forms Revision: Myeloma Changes

Case Report Membranoproliferative Glomerulonephritis in Patients with Chronic Venous Catheters: A Case Report and Literature Review

Glomerular Diseases. Davis Massey, MD, PhD Surgical Pathology Anna Vinnikova, MD Nephrology

Renal Pathology Case Conference. Case 2

Dense deposit disease with steroid pulse therapy

GRAND ROUNDS

Proliferative glomerulonephritis with monoclonal IgG deposits: A distinct entity mimicking immune-complex glomerulonephritis

29 Glomerular disease: an overview

Diabetes, Obesity and Heavy Proteinuria

Case # 2 3/27/2017. Disclosure of Relevant Financial Relationships. Clinical history. Clinical history. Laboratory findings

Favorable effect of bortezomib in dense deposit disease associated with monoclonal gammopathy: a case report

A Case of Myeloma Kidney With Glomerular C3 Deposition

Secondary IgA Nephropathy & HSP

Tuesday Conference 7/23/2013. Hasan Fattah

Instructions for Plasma Cell Disorders (PCD) Post-HCT Data (Form 2116 Revision 3)

An unusual association between focal segmental sclerosis and lupus nephritis: a distinct concept from lupus podocytopathy?

C3G An Update What is C3 Glomerulopathy Anyway? Patrick D. Walker, M.D. Nephropath Little Rock, Arkansas USA

Membranoproliferative Glomerulonephritis Secondary to Monoclonal Gammopathy

Membranoproliferative Glomerulonephritis

Dr Ian Roberts Oxford

Clinical pathological correlations in AKI

Glomerular Disease. January 16, Katharine Dahl, MD

Multiple intra-renal pathological injury patterns in resistant myeloma

Clinicopathologic Characteristics of IgA Nephropathy with Steroid-responsive Nephrotic Syndrome

Mr. I.K 58 years old

Lec-14 د.خالد نافع. Medicine. Multiple Myeloma

Transcription:

Hindawi Publishing Corporation Case Reports in Nephrology Volume 214, Article ID 164694, 6 pages http://dx.doi.org/1.1155/214/164694 Case Report Nephrotic Syndrome Secondary to Proliferative Glomerulonephritis with Monoclonal Immunoglobulin Deposits of Lambda Light Chain Seongseok Yun, 1,2 Beth L. Braunhut, 3 Courtney N. Walker, 1 Waheed Bhati, 1 Amy N. Sussman, 1 and Faiz Anwer 4 1 Department of Medicine, University of Arizona, Tucson, AZ 85721, USA 2 Department of Medicine, Arizona Health Sciences Center, 6th Floor, Room 6336, 151 N. Campbell Avenue,Tucson, AZ 85719, USA 3 Division of Pathology, University of Arizona, Tucson, AZ 85721, USA 4 Division of Hematology, Oncology, Blood & Marrow Transplantation, Department of Medicine, University of Arizona, Tucson, AZ 86721, USA Correspondence should be addressed to Seongseok Yun; namaska97@gmail.com Received 14 April 214; Revised 25 June 214; Accepted 7 July 214; Published 22 July 214 Academic Editor: Ricardo Enríquez Copyright 214 Seongseok Yun et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. We describe a rare case of a 46-year-old woman with history of refractory nephrotic syndrome and hypertension who presented with worsening proteinuria and kidney function. Work-up for both autoimmune and infectious diseases and hematologic malignancies including multiple myeloma were negative. Kidney biopsy demonstrated glomerular sclerotic change with lambda light chain deposits in the subendothelial space, which is consistent with proliferative glomerulonephritis with monoclonal immunoglobulin deposit (PGNMID). The patient was treated with bortezomib and dexamethasone without clinical improvement and eventually became hemodialysis dependent. 1. Introduction PGNMID is a recently described as a rare disorder that belongs to the class of disorders known as monoclonal gammopathy of renal significance (MGRS) [1]. PGNMID has been shown to be associated with various underlying diseases including myeloma, chronic lymphocytic leukemia (CLL), and parvovirus infection [1 5], and the most common clinical manifestation is renal involvement. PGNMID has unique histological and pathophysiological features, which distinguish it from other entities of MGRS or malignant monoclonal diseases. The renal deposit in PGNMID is composed of monoclonal immunoglobulin (Ig). Microscopic findings typically show a nonfibrillar or nonmicrotubular pattern of Ig deposit without any organized structure. Although histologic features have been well described and the clinical course of PGNMID is known to be poor, the underlying mechanisms of PGNMID remain elusive. Based on the evidence of other clonal disorders, bortezomib, cyclophosphamide, and high dose of chemotherapy followed by stem cell transplant are the current recommended treatments, although with variable response. 2. Case Presentation A 46-year-old woman with history of nephrotic syndrome was referred from an outside hospital for further evaluation and management of recurrent and progressively worsening nephrotic syndrome with stage III chronic kidney disease (CKD). Kidney biopsy and immunofluorescence (IF) staining one year ago had shown focal segmental proliferative glomerulonephritis and the patient had been treated with 16 weeks of cyclosporine followed by prednisone. Unfortunately, one year later, she presented with recurrent nephrotic symptoms including elevated serum creatinine, hypoalbuminemia, and significant proteinuria (Figure 1). Vital signs revealed blood pressure of 155/83 mmhg, heart rate of 81 beats per minute, respiratory rate of 14/minute,

2 Case Reports in Nephrology 9 9 8 8 8 Creatinine and phosphorus (mg/dl) 7 6 5 4 3 2 Bortezomib dexamethasone Dialysis 1 Cyclosporin prednisone 5 1 15 2 25 Month 6 3 BUN (mg/dl) Urine albumin and random protein (mg/dl) 6 4 2 Bortezomib dexamethasone Dialysis Cyclosporin prednisone 5 1 15 2 25 Month 6 4 2 Albumin/creatinine ratio (g/g) Creatinine BUN Phosphorus Albumin Albumin/creatinine Random protein Figure 1: Laboratory values during the chemotherapy. 16-week treatment with cyclosporine followed by prednisone failed to prevent disease progression. Patient received a total of 4 cycles of bortezomib and dexamethasone, however, with no clinical or laboratory improvement. Disease progressed and patient eventually became hemodialysis dependent. and temperature of 37.6 C. Lung and heart exams were normal and there was no palpable cervical, supraclavicular, axillary, or inguinal lymphadenopathy. Bilateral, one plus pitting lower extremity edema was present. Laboratory exam showed serum creatinine of 1.92 (normal.67 1.17 mg/dl) (Figure 1), IgG of 1333 (normal 552 1631 mg/dl), elevated kappa chain of 3.67 (normal.33 1.94 mg/dl), and lambda chain of 2.29 (normal.57 2.63 mg/dl) with normal ratio of 1.62. A 24-hour urine protein excretion was 8 g (normal 8 mg), and urine and serum immunofixation demonstrated positive monoclonal-free lambda light chain. Serum IgA (21 mg/dl), IgM (12 mg/dl), C3 (152 mg/dl), and C4 (32 mg/dl) levels were within normal range. Additional rheumatologic work-up included anti-ssa, anti-ssb, anti-nuclear antibodies (ANA), anti-neutrophil cytoplasmic antibody (ANCA), serum cryoglobulin, hepatitis panel, and serum protein electrophoresis (SPEP), which were all negative. Renal biopsy with light microscopic examination revealed multiple globally sclerosed glomeruli and occasional glomeruli with segmental sclerosis. The glomerular capillary loops showed patchy endocapillary proliferation and mononuclear inflammatory cell infiltration. A patchy mononuclear cell infiltrate was observed with associated tubulitis (Figures 2 and 2). Immunofluorescence (IF) examination revealed mesangial staining for IgG (trace) and IgM (trace) and glomerular capillary loops were stained positive for C3 (1 to 2+) and lambda light chain (2+) (Figures 3 3(d)). Subsequent electron microscopic ultrastructural examination demonstrated scattered electron dense immune-type deposits within mesangial areas and focally within the subendothelial space. The complexes did not show any organized substructural features, and the glomerular basement membrane had markedly irregular thickness in areas involved with deposits and/or endocapillary proliferation (Figures 4 and 4). Further work-up for multiple myeloma, including skeletal survey and bone marrow biopsy, was normal with only 5% plasma cells on bone marrow immunohistochemistry and only.6% CD2+ B cells on flow cytometry. Additional fluorescence in situ hybridization (FISH) analysis included 1q21 (CKS1B), 9q34 (ASS1), 11q13 (CCND1), 14q32 (IGH), 15q22 (PML), and 17p13 (TP53), which were all negative. Although the IF staining for IgG is weaker than that of previous case reports, suggesting either early stage of disease or suboptimal staining condition, the microscopic findings and laboratory results confirmed the diagnosis of proliferative glomerulonephritis with monoclonal immunoglobulin deposit (PGNMID) with monoclonal lambda light chain. The patient completed three cycles of weekly bortezomib along with dexamethasone without complication. Unfortunately, her disease progressed and she became hemodialysis dependent (Figure1). 3. Discussion PGNMID is a rare disease (incidence of.17%) with most published data extrapolated from case reports [1]. Proposed criteria to diagnosis PGNMID include (1) the presence of Ig

Case Reports in Nephrology 3 Figure 2: Light microscopic finding of kidney biopsy. PAS at 4x shows glomerulus with globally increased mesangial matrix and cellularity and segmental attachment to Bowman s capsule. The glomerular capillary loops show patchy endocapillary proliferation and focal mononuclear cell infiltration. No necrosis or crescents are seen in glomeruli, and the interstitium shows extensive fibrosis and patchy tubular atrophy. PAS at 2x demonstrates tubules containing intraluminal casts with smooth borders and flattening and sloughing of tubular epithelial cells. (c) (d) Figure 3: Immunofluorescence staining of kidney biopsy. Immunofluorescence staining shows trace mesangial staining for IgG, IgM, and kappa light chain (c) but 2+ mesangial and granular capillary loop staining for lambda light chain (d), confirming the diagnosis of PGNMID with monoclonal lambda light chain. deposit with a single IgG subclass and a single light chain isotype, (2) granular pattern of deposit under microscopy, and (3) no evidence of cryoglobulinemia [6]. PGNMID belongs to the disease spectrum known as monoclonal gammopathy of renal significance (MGRS) that includes amyloidosis (AL), type I cryoglobulinemia, immunotactoid glomerulopathy (ITG), and Randall-type monoclonal immunoglobulin deposition disease (MIDD) [7]. AL is one of the most frequent diseases in MGRS and is associated with plasma cell clones [6]. The deposit is composed of monoclonal light chain or its fragments. Although 7 8% of AL patients have renal involvement, the mortality largely depends on cardiac

4 Case Reports in Nephrology Figure 4: Electron microscopic finding of kidney biopsy. Electron microscopy demonstrates diffuse podocyte foot process effacement, proliferation in the capillary loop, and remodeling of the glomerular basement membrane with cellular interposition. The glomerular basement membrane is markedly irregular in thickness, and dense deposits are located in the mesangial areas and focally within subendothelial space without any organized substructural features. In loops without these abnormalities, the glomerular basement membrane is of normal thickness and has normal architecture. involvement. Type I cryoglobulinemia is another monoclonal disease with single monoclonal Ig deposit [6]. IgG type is the most common type and is precipitated by cold temperature. Raynaud s phenomenon, glomerular disease of membranoproliferative glomerulonephritis (MPGN), hypertension, and articular involvement are common clinical manifestations (Table 1). Randal-type MIDD is frequently associated with multiple myeloma with more than 1% of bone marrow involvement with plasma cells [6]. The most common form of Randal-type MIDD is light chain deposition disease (LCDD). Light heavy chain deposition disease (LHCDD) and heavy chain deposition disease (HCDD) are relatively rare. Glomerular deposits with nodular glomerulosclerosis and linear amorphous deposits in the tubular basement membranes are common histologic findings. Majority of the patients present with renal involvement including proteinuria, hematuria, and renal insufficiency. Renal biopsy and pathologic review are mandatory for the differential diagnosis of MGRS. B-cell clones that do not meet criteria for lymphoma or multiple myeloma are known to be responsible for MGRS. Most of the pathological and clinical consequences of MGRS arise from the deposition of monoclonal Ig in the organ rather than the proliferation of abnormal B-cell clones [8]. Moreover, PGNMID has a distinct microscopic appearance compared to other entities of MGRS, including a granular nonlinear pattern without deposits in glomerular or tubular basement membranes [1, 2, 6] and diffuse endocapillary proliferative glomerulonephritis (DPGN) or membranoproliferative glomerulonephritis (MPGN) pattern. Patients with PGNMID most commonly present with proteinuria, renal insufficiency, and hematuria without clinical evidence of multiple myeloma or B-cell lymphoproliferative disorders such as lymphadenopathy, hepatosplenomegaly, or skeletal involvement, and extrarenal Table 1: Clinical manifestations of PGNMID [1]. Pathologic findings Clinical findings Membranoproliferative Peripheral edema GN Endocapillary Hematuria proliferative GN Mesangial proliferative GN Membranous GN Focal interstitial inflammation Tubular atrophy and interstitial fibrosis Laboratory findings Proteinuria Hypoalbuminemia Renal insufficiency Serum and urine paraprotein Low C3 and C4 manifestations of PGNMID are extremely rare (Table 2). Additionally, the renal Ig deposits are not a consequence of glomerular damage secondary to autoimmune, infectious, or systemic disorders that can cause antigenic stimulation. Overall, the pathogenesis of PGNMID is poorly understood. Only one-third of patients showed a positive monoclonal spike in either SPEP or UPEP, and the plasma cells found in the bone marrow are usually less than 1% [1]. In our patient, serum kappa light chain was slightly elevated and lambda chain was normal with normal ratio of 1.62. However, monoclonal renal deposits indicate the presence of circulating monoclonal plasma cells and an underlying B-cell disorder as shown in our patient, and this has been the rationale for using chemotherapy targeting B-cells in PGNMID [9]. To date, there is no treatment that inhibits the process of Ig accumulation or removes already formed immune deposits. Currently, most therapies for PGNMID target

Case Reports in Nephrology 5 Table 2: Treatment recommendation for PGNMID [9]. CKD stage I and II Proteinuria (<1g/day) No evidence of progressive disease (1) Symptomatic measures only (2) Careful surveillance CKD stage I and II with high grade proteinuria (>1g/day) Progressive disease CKD stage III and IV (1) Chemotherapy, cyclophosphamide, and bortezomib (2) HDM/ASCT (3) Rituximab CKD stage V (1) Kidney transplantation (2) HDM/ASCT the underlying B-cell disorder using chemotherapeutic or immune-modulatory agents, although evidence is lacking. There are several case reports of PGNMID patients treated with chlorambucil, mycophenolate mofetil, prednisone, thalidomide, and rituximab, but the outcomes are disappointing with low remission rates [1, 4, 1]. The International Kidney and Monoclonal Gammopathy Research Group recommends treatment according to the severity of renal disease (Table 2) [9]. Symptomatic management isrecommendedforckdstageioriiwithproteinuria less than 1 g/day and no evidence of disease progression. Chemotherapy with bortezomib or cyclophosphamide is recommended for CKD stage I or II with proteinuria greater than 1 g/day and progressive disease. High dose melphalan followed by autologous stem cell transplant (HDM/ASCT) is an alternative option for patients less than 65 years of age. However, the efficacy of these chemotherapy targeting plasma cells is unknown especially in a setting of less than 1% plasma cells in PGNMID warranting further clinical investigation. Accordingly, rituximab targeting CD2+ B cell clones is excluded from current recommendations due to its limited efficacy and delayed complications [11], although it has been shown to be effective in several PGNMID cases [4]. Lastly, kidney transplantation with or without pretransplant chemotherapy or HDM/ASCT is the best suggested option for patients with CKD stage V. Following the recommendation of the International Kidney and Monoclonal Gammopathy Research Group, our patient was treated with bortezomib and dexamethasone, although without improvement. Rituximab was not consideredforthispatientduetolackofcd2+populationin flow cytometry in bone marrow cells. Eventually, the patient s renal disease progressed to hemodialysis dependence. Alternative treatment options are now HDM/ASCT or kidney transplant. As described in this case, there are several limitations in the current treatment guidelines. First of all, therapies are not basedonprospective,randomizedcontrolledclinicaltrials but on a limited number of case studies. Secondly, treatment simply depends on CKD staging without an understanding of the underlying pathophysiology of monoclonal production and deposition. Thirdly, no prognostic factor or prognostic system has been investigated or developed yet. Finally, more clinical studies are necessary to better understand PGNMID and improve clinical outcomes of this rare disease. 4. Conclusion PGNMID is a very rare disease of the MGRS spectrum. The pathologic abnormalities are limited to the renal system, and common clinical manifestations include proteinuria, hypoalbuminemia, and renal insufficiency. The underlying mechanisms of PGNMID remain elusive. However, the presence of monoclonal deposits in the kidney suggests an underlying plasma cell and B-cell disorder. Depending on the stageofckd,thecurrenttherapeuticoptionsarebortezomib, cyclophosphamide, hemodialysis, or stem cell transplant, although evidence is lacking. Conflict of Interests The authors declare that there is no conflict of interests regarding the publishing of this paper. References [1] S. H. Nasr, A. Satoskar, G. S. Markowitz et al., Proliferative glomerulonephritis with monoclonal IgG deposits, Journal of the American Society of Nephrology, vol.2,no.9,pp.255 264, 29. [2]E.Guiard,A.Karras,E.Plaisieretal., Patternsofnoncryoglobulinemic glomerulonephritis with monoclonal Ig deposits: correlation with IgG subclass and response to rituximab, Clinical Journal of the American Society of Nephrology, vol.6, no.7,pp.169 1616,211. [3] M.D.Redondo-Pachón, R. Enríquez, A. E. Sirvent et al., Proliferative glomerulonephritis with monoclonal IgG deposits in multiple myeloma, Nefrologia,vol.32,no.6,pp.846 848,212. [4]S.J.Barbour,M.C.Beaulieu,N.Y.Zalunardo,andA.B. Magil, Proliferative glomerulonephritis with monoclonal IgG deposits secondary to chronic lymphocytic leukemia. Report of two cases, Nephrology Dialysis Transplantation, vol.26,no.8, pp.2712 2714,211. [5] E. Fujita, A. Shimizu, T. Kaneko et al., Proliferative glomerulonephritis with monoclonal immunoglobulin G3κ deposits in association with parvovirus B19 infection, Human Pathology, vol.43,no.12,pp.2326 2333,212. [6] S. H. Nasr, G. S. Markowitz, M. B. Stokes et al., Proliferative glomerulonephritis with monoclonal IgG deposits: a distinct entity mimicking immune-complex glomerulonephritis, Kidney International,vol.65,no.1,pp.85 96,24. [7] N.Leung,F.Bridoux,C.A.Hutchisonetal., Monoclonalgammopathy of renal significance: when MGUS is no longer

6 Case Reports in Nephrology undetermined or insignificant, Blood,vol.12,no.22,pp.4292 4295, 212. [8] G. Merlini and M. J. Stone, Dangerous small B-cell clones, Blood,vol.18,no.8,pp.252 253,26. [9]J.P.Fermand,F.Bridoux,R.A.Kyleetal., HowItreat monoclonal gammopathy of renal significance (MGRS), Blood, vol. 122, pp. 3583 359, 213. [1] A. Ranghino, M. Tamagnone, M. Messina et al., A case of recurrent proliferative glomerulonephritis with monoclonal IgG deposits after kidney transplant treated with plasmapheresis, Case Reports in Nephrology and Urology, vol. 2, no. 1, pp. 46 52, 212. [11] C. Tarella, R. Passera, M. Magni et al., Risk factors for the development of secondary malignancy after high-dose chemotherapy and autograft, with or without rituximab: a 2- year retrospective follow-up study in patients with lymphoma, Journal of Clinical Oncology,vol.29,no.7,pp.814 824,211.