Skip to main content
Erschienen in: BMC Nephrology 1/2017

Open Access 01.12.2017 | Case report

Long-term renal survival of γ3-heavy chain deposition disease: a case report

verfasst von: Takayuki Katsuno, Shige Mizuno, Masatsuna Mabuchi, Naotake Tsuboi, Atsushi Komatsuda, Shoichi Maruyama

Erschienen in: BMC Nephrology | Ausgabe 1/2017

Abstract

Background

Monoclonal immunoglobulin deposition disease (MIDD) is characterized by the non-amyloid deposition of monoclonal immunoglobulin fragments in the basement membranes. Heavy chain deposition disease (HCDD) is a type of MIDD. HCDD is an extremely rare disease, and only three cases have been reported in Japan up to the present. The prognosis of HCDD is very poor, and optimal treatment has not been established. Only a few cases of HCDD with favorable long-term renal prognosis have been reported to date.

Case presentation

The authors describe a 61-year-old woman who presented with massive proteinuria, progressive kidney impairment, and hypocomplementemia. Kidney biopsy was performed for a precise diagnosis. On light microscopy, glomerules were lobulated and presented with nodular sclerosing glomerulopathy with membranoproliferative glomerulonephritis-like features. Immunofluorescence studies were positive for IgG, C3, and C1q within the mesangial nodules and in a linear distribution along the capillary walls without associated deposition of light chains. Staining for IgG showed the presence of linear deposits along tubular basement membranes. The analysis of the IgG subclass stain demonstrated intense positivity for IgG3 only. Electron microscopy revealed non-organized electron-dense deposits in the expanded mesangial area and inner aspect of the glomerular basement membranes. In accordance with the histological findings, we diagnosed γ3-HCDD. There was no evidence of plasma cell dyscrasia as a result of bone marrow aspiration. Serum and urine monoclonal proteins were not detected by immunoelectrophoresis and immunofixation electrophoresis. The serum free light chain ratio was within normal range. At first, prednisolone was administrated at a dose of 40 mg/day. However, a therapeutic effect was not observed. Urinary protein was not decreased and renal function further deteriorated. Therefore, melphalan plus prednisolone (MP) therapy was initiated. After 4 courses of MP therapy, the clinical parameters, including proteinuria, serum creatinine, albumin, and complement level (C3 and C4) were ameliorated. To date, the patient has been followed for 28 months, and long-term renal survival has been observed.

Conclusions

In this case, hematologic disease such as multiple myeloma was not detected; however, MP therapy was effective. Recently, the novel concept of monoclonal gammopathy of renal significance (MGRS) has been reported. MIDD, which includes HCDD, is one category of MGRS. In MGRS, aggressive chemotherapy may induce favorable renal outcomes.
Abkürzungen
CKD
Chronic kidney disease
EDD
Electron-dense deposits
FITC
Fluorescein isothiocyanate
GBM
Glomerular basement membranes
HCDD
Heavy chain deposition disease
LCDD
Light chain deposition disease
LHCDD
Light and heavy chain deposition disease
MGRS
Monoclonal gammopathy of renal significance
MIDD
Monoclonal immunoglobulin deposition disease
MM
Multiple myeloma
MP
Melphalan plus prednisolone
NS
Nephrotic syndrome
PSL
Prednisolone
TBM
Tubular basement membranes

Background

Monoclonal immunoglobulin deposition disease (MIDD) has three distinct forms: light chain deposition disease (LCDD), light and heavy chain deposition disease (LHCDD), and heavy chain deposition disease (HCDD). These three subtypes are distinguished depending on the components of the deposits. The deposits are derived from monoclonal light chains only in LCDD, monoclonal light and heavy chains in LHCDD, and monoclonal heavy chains only in HCDD. Renal involvement is frequent in MIDD. The main clinical features are renal dysfunction and proteinuria, often accompanied by nephrotic syndrome (NS). The characteristic pathological findings in MIDD are the presence of linear non-organized monoclonal immunoglobulin deposits along tubular basement membranes (TBM) and glomerular basement membranes (GBM) by immunofluorescence and electron microscopy. Especially in cases of HCDD, light microscopy shows nodular sclerosing glomerulopathy. Among cases of MIDD, HCDD is extremely rare. This rare disease was first described in 1992 [1]. To date, more than 20 years have elapsed from the first publication, only a small number of articles have been presented, and most of them are case reports [114]. In Japan, only three cases have been reported until now [2, 5, 11]. HCDD is classed into three subtypes (γ, α, and μ) depending on the components of the heavy chain deposits. IgG has four subclasses, and γ-HCDD is further classified into four subtypes (γ1–4). As a treatment option for MIDD including HCDD, effectiveness such as blood stem-cell autografting, alkylating agents, rituximab and thalidomide has been reported in the previous reports [7, 1416]. However, the optimal therapeutic method for MIDD remains clearly undefined, and many cases have shown poor outcomes [3, 8, 9, 1214, 17]. In HCDD, deletion of the first constant heavy chain domain (CH1) might be one possible pathogenesis [9, 14, 17, 18]. Deletion of the CH1 is a required condition for the secretion of a free monoclonal heavy chain by the underlying clonal plasma cell disorder [19]. Based on this pathogenesis, therapies targeting pathological plasma cell clones may be useful. Soma et al. and Oe et al. reported two cases of HCDD treated successfully using melphalan and prednisolone (MP) therapy [2, 5]. Several reports have shown beneficial results with high-dose melphalan followed by autologous stem cell transplantation [2023]. We herein describe a case of γ3-HCDD which demonstrated long-term renal survival after MP therapy. This is the fourth case in Japan.

Case presentation

A 61-year-old woman with a medical history of hypertension and chronic kidney disease (CKD) was admitted to our hospital for proteinuria and progressive deterioration of renal function. She attended the hospital as an outpatient and was administered olmesartan, cilnidipine, and azosemide. Her serum creatinine level was 1.0 mg/dL, and no urinary abnormalities were observed in the local clinic. 2 months before admission, there were subjective symptoms of pedal edema and blood pressure elevation. Her creatinine level increased to 1.52 mg/dL, and heavy proteinuria was observed at a local clinic. Subsequently, she was referred to our hospital for further management.
At the initial visit, the patient had a blood pressure of 135/88 mmHg, body height of 149.5 cm, and weight of 42.5 kg. Physical examination revealed bilateral edema of the lower extremities. Urinalysis showed proteinuria with protein excretion of 4.15 g/g creatinine and 5–8 red blood cells per high-power field. Hematologic tests indicated a white blood cell count of 6.3 × 103/μL, hemoglobin level of 12.0 g/dL, and platelet count of 315 × 103/μL. Laboratory investigation revealed a serum creatinine concentration of 1.85 mg/dL, blood urea nitrogen of 37 mg/dL, and estimated glomerular filtration rate of 22.8 mL/min/1.73 m2. The total serum protein level was 5.2 g/dL with a serum albumin level of 3.1 g/dL. The following laboratory data were obtained: total cholesterol: 280 mg/dL, low-density lipoprotein cholesterol: 175 mg/dL, triglycerides: 237 mg/dL, uric acid: 6.3 mg/dL, C-reactive protein: 0.03 mg/dL. Liver dysfunction, diabetes mellitus, and electrolyte imbalance were not observed. Immunological studies showed a serum IgG concentration of 283 mg/dL (normal range 870–1700 mg/dL), IgA of 259 mg/L (normal range 110–410 mg/dL), and IgM of 183 mg/dL (normal range 35–220 mg/dL). The serum complement C3 level was 33.2 mg/dL (normal range 86–160 mg/dL), C4 was 3.3 mg/dL (normal range 17-45 mg/dL), and total hemolytic activity (CH50) was 9.1 U/mL (normal range 31.6–57.6 U/mL). Marked hypocomplementemia was recognized. Tests for hepatitis B and C, rheumatoid factor, anti-nuclear antibody, anti-DNA antibody, anti-SS-A/SS-B antibody, anti-neutrophil cytoplasmic antibody, anti-cardiolipin antibody, and cryoglobulin were all negative.
She was admitted to our hospital 2 days after the initial consultation for a careful examination. As a result of the additional examinations, the serum and urine immunoelectrophoresis showed no monoclonal bands. Furthermore, the entity of monoclonal protein was not identified on immunofixation electrophoresis. The serum free κ light chain level was 45.1 mg/L, λ light chain was 57.6 mg/L, and κ/λ ratio was 0.783 (normal range 0.248–1.804). Serum amyloid A protein level was not elevated at 2.7 μg/mL (normal 0–8.0 μg/mL). In the bone marrow examination, the CD 138 positive cells proliferate slightly, but they were not clear tumorous proliferation. Plasma cells are 0.5% in the bone marrow and heteromorphy has not been confirmed. Pathological proliferation of plasma cells was also denied by flow cytometry. Ultrasound examination indicated renal atrophy; therefore, laparoscopic renal biopsy was performed for accurate diagnosis.

Renal biopsy

The kidney specimens were studied by light microscopy, immunofluorescence stain, and electron microscopy using standard techniques. For direct immunofluorescence stain, 3-μm cryostat sections were stained using polyclonal fluorescein isothiocyanate (FITC)-conjugated antibodies to IgG, IgA, IgM (Medical & Biological Laboratories, Nagoya, Japan), C3, C4, C1q, κ, and λ(Dako, Copenhagen, Denmark). Further staining of the IgG subclasses was performed using mouse monoclonal antibodies to human IgG1 (Bio-Rad Laboratories, California, USA), IgG2, IgG3 (Sigma-Aldrich, St. Louis, USA), and IgG4 (Binding Site, Birmingham, UK), followed by FITC-conjugated goat anti-mouse IgG (Jackson Immuno Research Inc., Pennsylvania, USA).
The sample contained 13 glomerules, none of which were globally sclerosed. By light microscopy, glomerules showed a diffuse increase in the mesangial matrix and mild mesangial hypercellularity. Most glomeruli were lobulated and exhibited remarkable nodular formation in the mesangial area that narrowed the capillary lumens. Capillary walls were irregularly thickened. Duplication of the GBM accompanied by endothelial cell swelling indicated an endothelium disorder. Endocapillary proliferation and mesangiolysis were observed in several glomeruli. Crescents were not detected in the specimen. Tubular atrophy and interstitial fibrosis was mild with patchy infiltration of mononuclear cells. Vessels showed moderate arteriosclerosis (Fig. 1). Congo red staining was negative. Immunofluorescence staining showed moderate-intensity (2+) for IgG and C3 and mild intensity (1+) for C1q in the mesangium and along the capillary walls. Staining for IgG also was positive on TBM. However, no staining for light chains was observed in the mesangium, along the capillary walls, or on the TBM. IgG subclasses showed intense staining for IgG3, but IgG1, IgG2, and IgG4 were entirely negative (Fig. 2). Electron microscopy revealed electron-dense deposits (EDD) without organized structures along the inner aspect of the GBM. In the expanded mesangium area, nodular EDD were observed (Fig. 3). The pathological findings in this case corresponded with γ3-HCDD.

Clinical course

Initially, the patient was treated with oral prednisolone (PSL) at a dose of 40 mg/day. However, the therapeutic response was very poor. The serum creatinine level increased to 3.65 mg/dL and blood urea nitrogen to 122 mg/dL, while the severe hypoalbuminemia (2.4 g/dL) did not improve. Urinary protein excretion did not decrease (4.27 g/g creatinine on admission to a maximum of 12.02 g/g creatinine), and in addition, the ascitic fluid and bilateral edema of the lower extremities became worse. The serum C3, C4, and CH50 titers remained extremely low (C3 19.7 mg/dL, C4 7.3 mg/dL, and CH50 13.6 U/mL). Therefore, the patient was treated with chemotherapy consisting of melphalan and PSL at a dose of 40 mg/day for 4 consecutive days. The daily use of PSL was rapidly tapered and stopped after chemotherapy. MP therapy was conducted 4 times in 7 months. The treatment interval was 1 month at short and 3 months at long. The dosage of melphalan was adjusted in accordance with renal function, 4 mg in the first and second courses, 6 mg in the third and 8 mg in the final. After 4 courses of MP therapy, the serum C3 and C4 titers were markedly elevated (C3 84.6 mg/dL and C4 31.3 mg/dL), and the serum creatinine level was decreased to 1.52 mg/dL. After discontinuance of MP therapy, the laboratory data did not deteriorate. To date, the patient has been followed for 28 months. The serum albumin levels have been maintained at over 3 g/dL, and the quantitative analysis of urinary protein was decreased to 2.72 g/g creatinine. NS was not observed and renal function was stable (the serum creatinine level was around 2.0 mg/dL). The patient’s condition improved with no edema, which had been observed initially.

Discussion

We report a case of γ-3HCDD which was diagnosed with clinicopathological features. Written informed consent was obtained from the patient for the administration of melphalan. She provided informed consent for the publication of this case report.
Kambham et al. described the disease spectrum of HCDD [17]. The most common clinical findings are NS, hypertension, microhematuria, and, in some cases, hypocomplementemia. Progressive renal dysfunction is usually present at the time of diagnosis. Lin et al. reported the clinicopathologic findings and outcomes of MIDD [18]. Of the 34 patients, 6 patients were diagnosed with HCDD. At baseline, the mean serum creatinine level was 4.8 ± 1.5 mg/dL (mean ± SEM: standard error of the mean), proteinuria was 5.3 ± 2.2 g/24 h, and the serum albumin level was 2.9 ± 0.3 g/dL. NS was seen in half of the patients with HCDD. Nasr et al. compared 64 patients with renal MIDD [15]. The mean 24-h urine protein was higher and NS occurrence was more frequent in patients with HCDD than LCDD. Oe et al. reported an overview of HCDD [24]. Extrarenal organ involvement of HCDD was observed in five cases, involving the skin, skeletal muscles, liver, and thyroid.
In our case, massive proteinuria and advanced renal failure were observed at the first visit. Organ damage was limited to the kidney. Severe hypocomplementemia was detected in our patient. The existence or nonexistence of hypocomplementemia is related to the IgG subclasses of HCDD. Soma et al. presented a first case report of γ3-HCDD in Japan and mentioned the relevance with hypocomplementemia and IgG subclasses [2]. In the previous cases, γ1 and γ3-HCDD cases showed hypocomplementemia. Particularly, severe hypocomplementemia with markedly low C3 and C4 levels were recognized in the γ3-HCDD cases. On the other hand, patients with γ2 and γ4-HCDD did not exhibit hypocomplementemia [1, 3]. Among IgG subclasses, IgG3 binds C1q most efficiently and strongly activates the classical pathway of the complement system [4]. In γ1 andγ3-HCDD, complement activation may be related enhanced C1q binding secondary to CH1 deletion. Therefore, in γ3-HCDD cases, including our patient, severely low C3 and C4 levels have been observed. In our case, immunofluorescence staining of the renal tissue revealed positivity of not only IgG, but also C3 and C1q. These findings did not contradict the classical pathway activation by IgG3. The serum complement levels correlated with the disease activity of HCDD. In previous cases, normalization of the serum complement titers with appropriate treatment was reported [5, 6].
The serum or urine monoclonal protein levels were positive in more than half of the HCDD patients (64–86% in the serum and 50–83% in the urine), but not all [24]. The absence of monoclonal protein is not sufficient to rule out HCDD. In our patient, routine serum and urine immunoelectrophoresis, serum immunofixation electrophoresis, and the serum free light chain ratio (κ /λ ratio) were examined for detection of monoclonal protein several times. However, serum and urine monoclonal protein was not found. In this case, bone marrow aspirate did not show monoclonal proliferation of plasma cells. In the previous article, a definitive diagnosis of plasma cell dyscrasia could be made in only 19% of cases of γ-HCDD [7].
Typical pathological findings of HCDD show nodular mesangial sclerosis. The mesangial nodules were generally positive for periodic acid-Schiff, trichrome blue, and silver stain. Crescentic formation is also sometimes observed [7]. Vedder et al. reported an unusual case of endocapillary proliferative glomerulonephritis due to HCDD [8]. An immunofluorescence study revealed linear positive staining on GBM and TBM for a single heavy chain with completely negative staining for light chains. In the majority of γ3-HCDD cases, complement deposition is detected. On electron microscopy, fine granular EDD along the inner aspect of the lamina densa of GBM and the outer aspect of the TBM were observed. EDD were also seen in the expanded mesangial area. Mesangial deposits tend to be more common in HCDD than LCDD [15].
The diagnosis of HCDD requires characteristic pathological findings. The following criteria were applied in previous cohort studies of MIDD; (A) positive staining for a single class of immunoglobulin (γ, α, μ) on the GBM and/or TBM, (B) completely negative staining of light chains (κ and λ), and (C) characteristic EDD observed by electron microscopy [15, 18, 24]. Renal pathological findings of this present case fulfilled these above criteria. The finding that only IgG3 is linearly positive for TBM was of particular importance. Although Aucouturier et al. suggested the hallmark of HCDD is CH1 deletion of γ-heavy chains [9], staining for CH1 is not necessarily required for a diagnosis. In addition to the pathological diagnosis, clinical features, which include NS with progressive renal failure, hypertension, and prominent hypocomplementemia, coincided with the previously reported clinical presentations.
The treatment of HCDD has not been established until now. The prognosis of HCDD is very poor historically. Kambham et al. reported 11 cases of HCDD treated with corticosteroids, cyclophosphamide, and melphalan [17]. In the 8 patients for whom follow-up data was available, 2 progressed to end-stage kidney disease, which required renal replacement therapy within less than 1 year, and 3 had worsening proteinuria and deterioration in renal function. Oe et al. reviewed the clinical outcomes and prognosis in a previous report [24]. Most HCDD patients are refractory to treatment. Within a year, 36–50% of patients develop end-stage kidney disease requiring hemodialysis. Within a mean duration of 12–76 months after onset, the mortality is about 10%. However, chemotherapy based on the regimen used for multiple myeloma (MM) was useful in some cases. Soma et al. and Oe et al. reported two cases of HCDD treated successfully using MP therapy [2, 5]. Yin et al. described a unique type of γ3-HCDD that achieved complete remission according to urinalysis by treatment with thalidomide and dexamethasone [10]. Royer et al. treated 11 young patients with MIDD by high dose chemotherapy with the support of autologous blood stem cell transplantation and reported the therapeutic benefit [20].
Recently, the utility of bortezomib has been reported. Patel et al. presented 3 patients with severe kidney manifestation of HCDD who achieved clinical remission and improvement of renal function following a bortezomib-based treatment [25]. Cohen et al. retrospectively studied 49 patients with MIDD who received intravenous bortezomib plus dexamethasone. In many patients studied, hematological and renal response was achieved. Bortezomib-based therapy is considered to be beneficial mainly in MIDD cases diagnosed earlier [26]. Bridoux et al. researched 15 patients with biopsy-proven HCDD, retrospectively. All patients received chemotherapy, including bortezomib in 10 cases. Renal clinical manifestation ameliorated in 11 patients who achieved a hematological response. In this report, the importance of early diagnosis and bortezomib-based treatment is mentioned for the preservation of renal prognosis [19]. In bortezomib-based therapy, serum free light chain response is consider to be a favorable prognostic factor for renal survival [27].
Therapeutic indicators for treating HCDD are urinalysis findings and renal function. Particularly, the serum complement level is important in γ3-HCDD patients. In our case, treatment with steroids alone had no effect at all. However, after the treatment with MP therapy, the serum albumin levels have been maintained at over 3.0 g/dL, and NS was not observed for a long period of time. The serum creatinine value was stable.
Hypocomplementemia was notably improved. For these reasons, MP therapy was considered to be beneficial in this case. Interestingly, our patient responded well to the MP therapy, although a bone marrow aspirate did not show findings of plasm cell infiltration, and monoclonal immunoglobin was not identified in the serum or urine by conventional electrophoresis and immunofixation. These results indicate that the monoclonal immunoglobin levels are very small, reflecting a strong affinity for kidney tissues. In fact, in this case, monoclonal immunoglobin was demonstrated only in a renal biopsy by immunofluorescence staining. The initiation of therapeutic intervention at an early stage of disease was indicated as a favorable prognostic factor in previous reports [2, 5, 15, 18, 24]. The response to MP therapy in this case may be interpreted by the early diagnosis. On histological findings, mild chronic changes such as tubular atrophy, interstitial fibrosis, and the absence of glomerular sclerosis lesions were also considered to be related to the treatment response.
In recent years, a review of monoclonal gammopathy of renal significance (MGRS) has been reported [28]. MGRS is a disease group of renal impairment induced by a monoclonal protein. MGRS is a new concept, and in 2012 the International Kidney and Monoclonal Gammopathy Research Group refined the definition of MGRS to distinguish these monoclonal gammopathies from monoclonal gammopathy of undetermined significance [29]. MIDD, including HCDD, is one category of MGRS. The monoclonal protein may cause kidney injury directly through non-organized deposits in MIDD. In MGRS, the amount of monoclonal protein and rapid deposition in the kidney influence the failure to detect monoclonal protein in the serum and urine. Therefore, kidney biopsy is needed for diagnosis of MGRS. In this case, the renal pathology findings were most important for the diagnosis of HCDD. Particularly, no immunofluorescence staining for light chains was a significant clue. In this case, although MM was not detected, chemotherapy based on the treatment regimen for MM was effective. More recently, it has been reported on the management of MGRS [30]. In this article, the treatment with high-dose melphalan supported by autologous peripheral blood stem cell transplantation and bortezomib-based regimens is recommended for the MIDD patients. In MGRS, early recognition and aggressive adaptation of the chemotherapeutic regimen may be crucial for renal prognosis.

Conclusions

In conclusion, we present a case of γ3-HCDD with NS and renal dysfunction who responded to MP therapy and maintained kidney function for a long time. However, further careful observation is necessary to assess the treatment effect. In order to improve the renal outcome of HCDD, an excellent understanding of renal pathology for early diagnosis and suitable treatment methods are required. Penetration of MGRS concept and aggressive chemotherapy may improve the renal prognosis.

Acknowledgements

The authors appreciate the excellent technical assistance of N. Asano, Y. Sawa, and N. Suzuki.

Funding

This work was supported by a Grant-in-Aid for Progressive Renal Diseases Research, Research on Intractable Disease, from the Ministry of Health, Labour, and Welfare of Japan.

Availability of data and materials

All data supporting the case are included in the manuscript.
Not applicable.
The patient provided informed consent for the publication of this case report.

Competing interests

The authors declare that they have no competing interests involving this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Tubbs RR, Berkley V, Valenzuela R, McMahon JT, Gephardt GN, Fishleder AJ, Nally JV, Pohl MA, Bukowski RM, Lichtin AE. Pseudo-Gamma heavy chain (IgG4 lambda) deposition disease. Mod Pathol. 1992;5:185–90.PubMed Tubbs RR, Berkley V, Valenzuela R, McMahon JT, Gephardt GN, Fishleder AJ, Nally JV, Pohl MA, Bukowski RM, Lichtin AE. Pseudo-Gamma heavy chain (IgG4 lambda) deposition disease. Mod Pathol. 1992;5:185–90.PubMed
2.
Zurück zum Zitat Soma J, Sato K, Sakuma T, Saito H, Sato H, Sato T, Abbas A, Aucouturier P. Immunoglobulin gamma 3-heavy-chain deposition disease: report of a case and relationship with hypocomplementemia. Am J Kidney Dis. 2004;43:E10–6.CrossRefPubMed Soma J, Sato K, Sakuma T, Saito H, Sato H, Sato T, Abbas A, Aucouturier P. Immunoglobulin gamma 3-heavy-chain deposition disease: report of a case and relationship with hypocomplementemia. Am J Kidney Dis. 2004;43:E10–6.CrossRefPubMed
3.
Zurück zum Zitat Herzenberg AM, Kiaii M, Magil AB. Heavy chain deposition disease: recurrence in a renal transplant and report of IgG2 subtype. Am J Kidney Dis. 2000;35:E25.CrossRefPubMed Herzenberg AM, Kiaii M, Magil AB. Heavy chain deposition disease: recurrence in a renal transplant and report of IgG2 subtype. Am J Kidney Dis. 2000;35:E25.CrossRefPubMed
4.
Zurück zum Zitat Meletic VD, Frank MM. Complement-immunoglobulin interactions. Curr Opin Immunol. 1995;7:41–7.CrossRef Meletic VD, Frank MM. Complement-immunoglobulin interactions. Curr Opin Immunol. 1995;7:41–7.CrossRef
5.
Zurück zum Zitat Oe Y, Nakaya I, Yahata M, Sakuma T, Sato H, Soma J. A case of γ1-heavy chain deposition disease successfully treated with melphalan and prednisolone therapy. Intern Med. 2010;49:1411–5.CrossRefPubMed Oe Y, Nakaya I, Yahata M, Sakuma T, Sato H, Soma J. A case of γ1-heavy chain deposition disease successfully treated with melphalan and prednisolone therapy. Intern Med. 2010;49:1411–5.CrossRefPubMed
6.
Zurück zum Zitat Soma J, Tsuchiya Y, Sakuma T, Sato H. Clinical remission and histopathological resolution of nodular lesions in a patient with gamma3 heavy-chain deposition disease. Clin Nephrol. 2008;69:383–6.CrossRefPubMed Soma J, Tsuchiya Y, Sakuma T, Sato H. Clinical remission and histopathological resolution of nodular lesions in a patient with gamma3 heavy-chain deposition disease. Clin Nephrol. 2008;69:383–6.CrossRefPubMed
7.
Zurück zum Zitat Alexander MP, Nasr SH, Watson DC, Méndez GP, Rennke HG. Renal crescentic alpha heavy chain deposition disease: a report of 3 cases and review of the literature. Am J Kidney Dis. 2011;58:621–5.CrossRefPubMed Alexander MP, Nasr SH, Watson DC, Méndez GP, Rennke HG. Renal crescentic alpha heavy chain deposition disease: a report of 3 cases and review of the literature. Am J Kidney Dis. 2011;58:621–5.CrossRefPubMed
8.
Zurück zum Zitat Vedder AC, Weening JJ, Krediet RT. Intracapillary proliferative glomerulonephritis due to heavy chain deposition disease. Nephrol Dial Transplant. 2004;19:1302–4.CrossRefPubMed Vedder AC, Weening JJ, Krediet RT. Intracapillary proliferative glomerulonephritis due to heavy chain deposition disease. Nephrol Dial Transplant. 2004;19:1302–4.CrossRefPubMed
9.
Zurück zum Zitat Aucouturier P, Khamlichi AA, Touchard G, Justrabo E, Cogne M, Chauffert B, Martin F, Preud’homme JL. Brief report: heavy-chain deposition disease. N Engl J Med. 1993;329:1389–93.CrossRefPubMed Aucouturier P, Khamlichi AA, Touchard G, Justrabo E, Cogne M, Chauffert B, Martin F, Preud’homme JL. Brief report: heavy-chain deposition disease. N Engl J Med. 1993;329:1389–93.CrossRefPubMed
10.
Zurück zum Zitat Yin G, Wu Y, Zeng CH, Chen HP, Liu ZH. A unique case of γ-heavy chain deposition disease characterized by concomitant deposition of γ2 and γ4 subclasses. Intern Med. 2014;53:2615–8.CrossRefPubMed Yin G, Wu Y, Zeng CH, Chen HP, Liu ZH. A unique case of γ-heavy chain deposition disease characterized by concomitant deposition of γ2 and γ4 subclasses. Intern Med. 2014;53:2615–8.CrossRefPubMed
11.
Zurück zum Zitat Yasuda T, Fujita K, Imai H, Morita K, Nakamoto Y, Miura AB. Gamma-heavy chain deposition disease showing nodular glomerulosclerosis. Clin Nephrol. 1995;44:394–9.PubMed Yasuda T, Fujita K, Imai H, Morita K, Nakamoto Y, Miura AB. Gamma-heavy chain deposition disease showing nodular glomerulosclerosis. Clin Nephrol. 1995;44:394–9.PubMed
12.
Zurück zum Zitat Katz A, Zent R, Bargman JM. IgG heavy-chain deposition disease. Mod Pathol. 1994;7:874–8.PubMed Katz A, Zent R, Bargman JM. IgG heavy-chain deposition disease. Mod Pathol. 1994;7:874–8.PubMed
13.
Zurück zum Zitat Herzenberg AM, Lien J, Magil AB. Monoclonal heavy chain (immunoglobulin G3) deposition disease: report of a case. Am J Kidney Dis. 1996;28:128–31.CrossRefPubMed Herzenberg AM, Lien J, Magil AB. Monoclonal heavy chain (immunoglobulin G3) deposition disease: report of a case. Am J Kidney Dis. 1996;28:128–31.CrossRefPubMed
14.
Zurück zum Zitat Moulin B, Deret S, Mariette X, Kourilsky O, Imai H, Dupouet L, Marcellin L, Kolb I, Aucouturier P, Brouet JC, Ronco PM, Mougenot B. Nodular glomerulosclerosis with deposition of monoclonal immunoglobulin heavy chains lacking C (H)1. J Am Soc Nephrol. 1999;10:519–28.PubMed Moulin B, Deret S, Mariette X, Kourilsky O, Imai H, Dupouet L, Marcellin L, Kolb I, Aucouturier P, Brouet JC, Ronco PM, Mougenot B. Nodular glomerulosclerosis with deposition of monoclonal immunoglobulin heavy chains lacking C (H)1. J Am Soc Nephrol. 1999;10:519–28.PubMed
15.
Zurück zum Zitat Nasr SH, Valeri AM, Cornell LD, Fidler ME, Sethi S, D’Agati VD, Leung N. Renal monoclonal immunoglobulin deposition disease: a report of 64 patients from a single institution. Clin J Am Soc Nephrol. 2012;7:231–9.CrossRefPubMed Nasr SH, Valeri AM, Cornell LD, Fidler ME, Sethi S, D’Agati VD, Leung N. Renal monoclonal immunoglobulin deposition disease: a report of 64 patients from a single institution. Clin J Am Soc Nephrol. 2012;7:231–9.CrossRefPubMed
16.
Zurück zum Zitat Kuypers DR, Lerut E, Claes K, Evenepoel P, Vanrenterghem Y. Recurrence of light chain deposit disease after renal allograft transplantation: potential role of rituximab? Transpl Int. 2007;20:381–5.CrossRefPubMed Kuypers DR, Lerut E, Claes K, Evenepoel P, Vanrenterghem Y. Recurrence of light chain deposit disease after renal allograft transplantation: potential role of rituximab? Transpl Int. 2007;20:381–5.CrossRefPubMed
17.
Zurück zum Zitat Kambham N, Markowitz GS, Appel GB, Kleiner MJ, Aucouturier P, D’Agati VD. Heavy chain deposition disease: the disease spectrum. Am J Kidney Dis. 1999;33:954–62.CrossRefPubMed Kambham N, Markowitz GS, Appel GB, Kleiner MJ, Aucouturier P, D’Agati VD. Heavy chain deposition disease: the disease spectrum. Am J Kidney Dis. 1999;33:954–62.CrossRefPubMed
18.
Zurück zum Zitat Lin J, Markowitz GS, Valeri AM, Kambham N, Sherman WH, Appel GB, D’Agati VD. Renal monoclonal immunoglobulin deposition disease: the disease spectrum. J Am Soc Nephrol. 2001;12:1482–92.PubMed Lin J, Markowitz GS, Valeri AM, Kambham N, Sherman WH, Appel GB, D’Agati VD. Renal monoclonal immunoglobulin deposition disease: the disease spectrum. J Am Soc Nephrol. 2001;12:1482–92.PubMed
19.
Zurück zum Zitat Bridoux F, Javaugue V, Bender S, Leroy F, Aucouturier P, Debiais-Delpech C, Goujon JM, Quellard N, Bonaud A, Clavel M, Trouillas P, Di Meo F, Gombert JM, Fermand JP, Jaccard A, Cogne ’ M, Touchard G, Sirac C. Unravelling the immunopathological mechanisms of heavy chain deposition disease with implications for clinical management. Kidney Int. 2017;91:423–34.CrossRefPubMed Bridoux F, Javaugue V, Bender S, Leroy F, Aucouturier P, Debiais-Delpech C, Goujon JM, Quellard N, Bonaud A, Clavel M, Trouillas P, Di Meo F, Gombert JM, Fermand JP, Jaccard A, Cogne ’ M, Touchard G, Sirac C. Unravelling the immunopathological mechanisms of heavy chain deposition disease with implications for clinical management. Kidney Int. 2017;91:423–34.CrossRefPubMed
20.
Zurück zum Zitat Royer B, Arnulf B, Martinez F, Roy L, Flageul B, Etienne I, Ronco P, Brouet JC, Fermand JP. High dose chemotherapy in light chain or light and heavy chain deposition disease. Kidney Int. 2004;65:642–8.CrossRefPubMed Royer B, Arnulf B, Martinez F, Roy L, Flageul B, Etienne I, Ronco P, Brouet JC, Fermand JP. High dose chemotherapy in light chain or light and heavy chain deposition disease. Kidney Int. 2004;65:642–8.CrossRefPubMed
21.
Zurück zum Zitat Firkin F, Hill PA, Dwyer K, Gock H. Reversal of dialysis-dependent renal failure in light-chain deposition disease by autologous peripheral blood stem cell transplantation. Am J Kidney Dis. 2004;44:551–5.CrossRefPubMed Firkin F, Hill PA, Dwyer K, Gock H. Reversal of dialysis-dependent renal failure in light-chain deposition disease by autologous peripheral blood stem cell transplantation. Am J Kidney Dis. 2004;44:551–5.CrossRefPubMed
22.
Zurück zum Zitat Telio D, Shepherd J, Forrest D, Zypchen L, Barnett M, Nevill T, Song KW. High-dose melphalan followed by auto-SCT has favorable safety and efficacy in selected patients with light chain deposition disease and light and heavy chain deposition disease. Bone Marrow Transplant. 2012;47:453–5.CrossRefPubMed Telio D, Shepherd J, Forrest D, Zypchen L, Barnett M, Nevill T, Song KW. High-dose melphalan followed by auto-SCT has favorable safety and efficacy in selected patients with light chain deposition disease and light and heavy chain deposition disease. Bone Marrow Transplant. 2012;47:453–5.CrossRefPubMed
23.
Zurück zum Zitat Brioli A, Zamagni E, Pasquali S, Tosi P, Tacchetti P, Perrone G, Pantani L, Petrucci A, Zannetti BA, Baccarani M, Cavo M. Long-term follow-up after autologous stem cell transplantation for light- and heavy-chain deposition disease. Bone Marrow Transplant. 2012;47:1248–9.CrossRefPubMed Brioli A, Zamagni E, Pasquali S, Tosi P, Tacchetti P, Perrone G, Pantani L, Petrucci A, Zannetti BA, Baccarani M, Cavo M. Long-term follow-up after autologous stem cell transplantation for light- and heavy-chain deposition disease. Bone Marrow Transplant. 2012;47:1248–9.CrossRefPubMed
24.
Zurück zum Zitat Oe Y, Soma J, Sato H, Ito S. Heavy chain deposition disease: an overview. Yuji Oe, Jun Soma, Hiroshi Sato, Sadayoshi Ito. Clin Exp Nephrol. 2013;17:771–8.CrossRefPubMed Oe Y, Soma J, Sato H, Ito S. Heavy chain deposition disease: an overview. Yuji Oe, Jun Soma, Hiroshi Sato, Sadayoshi Ito. Clin Exp Nephrol. 2013;17:771–8.CrossRefPubMed
25.
Zurück zum Zitat Patel K, Dillon JJ, Leung N, Bomback AS, Appel GB, D'Agati V, Canetta PA. Use of bortezomib in heavy-chain deposition disease: a report of 3 cases. Am J Kidney Dis. 2014;64:123–7.CrossRefPubMed Patel K, Dillon JJ, Leung N, Bomback AS, Appel GB, D'Agati V, Canetta PA. Use of bortezomib in heavy-chain deposition disease: a report of 3 cases. Am J Kidney Dis. 2014;64:123–7.CrossRefPubMed
26.
Zurück zum Zitat Cohen C, Royer B, Javaugue V, Szalat R, Karoui KE, Caulier A, Knebelmann B, Jaccard A, Chevret S, Touchard G, Fermand JP, Arnulf B, Bridoux F. Bortezomib produces high hematological response rates with prolonged renal survival in monoclonal immunoglobulin deposition disease. Kidney Int. 2015;88:1135–43.CrossRefPubMed Cohen C, Royer B, Javaugue V, Szalat R, Karoui KE, Caulier A, Knebelmann B, Jaccard A, Chevret S, Touchard G, Fermand JP, Arnulf B, Bridoux F. Bortezomib produces high hematological response rates with prolonged renal survival in monoclonal immunoglobulin deposition disease. Kidney Int. 2015;88:1135–43.CrossRefPubMed
27.
Zurück zum Zitat Cohen C, Javaugue V, Joly F, Arnulf B, Fermand JP, Jaccard A, Sirac C, Knebelmann B, Bridoux F, Touchard G. Randall-type monoclonal immunoglobulin deposition disease: From diagnosis to treatment. Nephrol Ther. 2016;12:131–9.CrossRefPubMed Cohen C, Javaugue V, Joly F, Arnulf B, Fermand JP, Jaccard A, Sirac C, Knebelmann B, Bridoux F, Touchard G. Randall-type monoclonal immunoglobulin deposition disease: From diagnosis to treatment. Nephrol Ther. 2016;12:131–9.CrossRefPubMed
28.
Zurück zum Zitat Bridoux F, Leung N, Hutchison CA, Touchard G, Sethi S, Fermand JP, Picken MM, Herrera GA, Kastritis E, Merlini G, Roussel M, Fervenza FC, Dispenzieri A, Kyle RA, Nasr SH. International kidney and monoclonal Gammopathy Research group. Diagnosis of monoclonal gammopathy of renal significance. Kidney Int. 2015;87:698–711.CrossRefPubMed Bridoux F, Leung N, Hutchison CA, Touchard G, Sethi S, Fermand JP, Picken MM, Herrera GA, Kastritis E, Merlini G, Roussel M, Fervenza FC, Dispenzieri A, Kyle RA, Nasr SH. International kidney and monoclonal Gammopathy Research group. Diagnosis of monoclonal gammopathy of renal significance. Kidney Int. 2015;87:698–711.CrossRefPubMed
29.
Zurück zum Zitat Leung N, Bridoux F, Hutchison CA, Nasr SH, Cockwell P, Fermand JP, Dispenzieri A, Song KW, Kyle RA. International kidney and monoclonal Gammopathy Research group. Monoclonal gammopathy of renal significance: when MGUS is no longer undetermined or insignificant. Blood. 2012;120:4292–5.CrossRefPubMed Leung N, Bridoux F, Hutchison CA, Nasr SH, Cockwell P, Fermand JP, Dispenzieri A, Song KW, Kyle RA. International kidney and monoclonal Gammopathy Research group. Monoclonal gammopathy of renal significance: when MGUS is no longer undetermined or insignificant. Blood. 2012;120:4292–5.CrossRefPubMed
30.
Zurück zum Zitat Fermand JP, Bridoux F, Kyle R, Kastritis E, Weiss B, Cook M, Drayson MT, Dispenzieri A, Leung N. How I treat monoclonal gammopathy of renal significance (MGRS). Blood. 2013;122:3583–90.CrossRefPubMed Fermand JP, Bridoux F, Kyle R, Kastritis E, Weiss B, Cook M, Drayson MT, Dispenzieri A, Leung N. How I treat monoclonal gammopathy of renal significance (MGRS). Blood. 2013;122:3583–90.CrossRefPubMed
Metadaten
Titel
Long-term renal survival of γ3-heavy chain deposition disease: a case report
verfasst von
Takayuki Katsuno
Shige Mizuno
Masatsuna Mabuchi
Naotake Tsuboi
Atsushi Komatsuda
Shoichi Maruyama
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2017
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-017-0645-z

Weitere Artikel der Ausgabe 1/2017

BMC Nephrology 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.