2,275
Views
23
CrossRef citations to date
0
Altmetric
Review

Management of myeloma-associated renal dysfunction in the era of novel therapies

, , , , , , , , & show all
Pages 51-68 | Published online: 10 Jan 2014

Abstract

Multiple myeloma (MM) is a plasma cell neoplasm often associated with renal impairment (RI), with myeloma cast nephropathy recognized as the most common cause. While RI is present in over 50% of MM patients at some point in their disease course, it is associated with higher tumor burden, more aggressive disease, diminished quality of life, development of complications and increased mortality. The introduction of novel therapies, including bortezomib, lenalidomide and thalidomide, has revolutionized the management of MM. They are now considered first-line therapies in induction, maintenance and salvage therapy for MM. In addition to their anti-MM effect, they can improve outcome in patients with RI, especially when combined, and bortezomib with dexamethasone may have a renal protective effect. This review focuses on the use of these agents in patients with MM and RI, and evaluates their efficacy, safety, need for dose adjustment and impact on RI.

Medscape: Continuing Medical Education Online

This activity has been planned and implemented in accordance with the Essential Areas and policies of the Accreditation Council for Continuing Medical Education through the joint sponsorship of Medscape, LLC and Expert Reviews Ltd. Medscape, LLC is accredited by the ACCME to provide continuing medical education for physicians.

Medscape, LLC designates this Journal-based CME activity for a maximum of 1 AMA PRA Category 1 Credit(s). Physicians should claim only the credit commensurate with the extent of their participation in the activity.

All other clinicians completing this activity will be issued a certificate of participation. To participate in this journal CME activity: (1) review the learning objectives and author disclosures; (2) study the education content; (3) take the post-test with a 70% minimum passing score and complete the evaluation at www.medscape.org/journal/experthematology; (4) view/print certificate.

Release date: January 23, 2012; Expiration date: January 23, 2013

Learning objectives

Upon completion of this activity, participants will be able to:

  • • Analyze the diagnostic process of MM

  • • Distinguish the predominant renal histologic subtype in cases of RI associated with MM

  • • Analyze treatment options for MM in the setting of RI

Financial & competing interests disclosure

EDITOR

Elisa Manzotti

Editorial Director, Future Science Group, London, UK.

Disclosure: Elisa Manzotti has disclosed no relevant financial relationships.

CME AUTHOR

Charles P Vega, MD

Health Sciences Clinical Professor; Residency Director, Department of Family Medicine, University of California, Irvine, CA, USA.

Disclosure: Charles P Vega has disclosed no relevant financial relationships.

AUTHORS AND CREDENTIALS

Mahmoud R Gaballa, MD

Department of Internal Medicine, Thomas Jefferson University, Philadelphia, PA, USA.

Disclosure: Mahmoud R Gaballa has disclosed no relevant financial relationships.

Jacob L Laubach, MD

Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.

Disclosure: Jacob L Laubach has disclosed no relevant financial relationships.

Robert L Schlossman, MD

Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.

Disclosure: Robert L Schlossman has disclosed no relevant financial relationships.

Katherine Redman

Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.

Disclosure: Katherine Redman has disclosed no relevant financial relationships.

Kimberly Noonan, NP

Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.

Disclosure: Kimberly Noonan has disclosed no relevant financial relationships.

Constantine S Mitsiades, MD, PhD

Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.

Disclosure: Constantine S Mitsiades has disclosed the following relevant financial relationships: he has received funding for clinical research from: Amgen Inc.; AVEO Pharma; Genzyme Corporation; and Johnson & Johnson Pharmaceutical Research & Development, LLC. He has served as a consultant or received honoraria from: Bristol-Myers Squibb Company; Celgene Corporation; Centocor Research & Development, Inc.; Merck & Co., Inc.; Millennium Pharmaceuticals, Inc.; and Novartis Pharmaceuticals Corporation.

Irene M Ghobrial, MD

Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.

Disclosure: Irene M Ghobrial has disclosed the following relevant financial relatioships: she has served on advisory boards for: Bristol-Myers Squibb Company; Genzyme Corporation; Millennium Pharmaceuticals, Inc.; Novartis Pharmaceuticals Corporation; and Onyx Pharmaceuticals, Inc.

Nikhil Munshi, MD

Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.

Disclosure: Nikhil Munshi has disclosed the following relevant financial relationships: he has served as a consultant for: Celgene Corporation; Millennium Pharmaceuticals, Inc.; Novartis Pharmaceuticals Corporation; and Onyx Pharmaceuticals, Inc.

Kenneth C Anderson, MD

Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.

Disclosure: Kenneth C Anderson has disclosed the following relevant financial relationships: he has served on advisory boards for: Bristol-Myers Squibb Company; Celgene Corporation; Merck & Co. Inc.; Millennium Pharmaceuticals, Inc.; Novartis Pharmaceuticals Corporation; and Onyx Pharmaceuticals, Inc.

Paul G Richardson, MD

Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.

Disclosure: Paul G Richardson has disclosed the following relevant financial relationships: he has served on advisory boards for: Bristol-Myers Squibb Company; Celgene Corporation; Johnson & Johnson Pharmaceutical Research & Development, LLC; Millennium Pharmaceuticals, Inc.; and Novartis Pharmaceuticals Corporation.

Figure 1. A simplified diagram illustrating the development of myeloma cast nephropathy.

Pathogenesis includes: (1) serum-free light chains produced in excess, overwhelming absorption mechanisms in the proximal tubule; (2) Bence–Jones proteins develop as a consequence of light chains entering the distal tubules; (3) Tamm–Horsfall proteins interact with Bence–Jones proteins in the distal tubule; (4) tubular cast formation and obstruction develop as the end product, with subsequent ‘myeloma cast nephropathy’ Citation[23].

Figure 1. A simplified diagram illustrating the development of myeloma cast nephropathy.Pathogenesis includes: (1) serum-free light chains produced in excess, overwhelming absorption mechanisms in the proximal tubule; (2) Bence–Jones proteins develop as a consequence of light chains entering the distal tubules; (3) Tamm–Horsfall proteins interact with Bence–Jones proteins in the distal tubule; (4) tubular cast formation and obstruction develop as the end product, with subsequent ‘myeloma cast nephropathy’ Citation[23].
Figure 2. A simplified diagram illustrating clinical manifestations and complications of multiple myeloma related to excess production of free light chains.

Excess production of free light chains is responsible for tubular cast formation and obstruction (myeloma cast nephropathy), induction of proinflammatory cytokines and consequent direct tubular cell injury, functional impairment (Fanconi syndrome) and a state of chronic inflammation through interference with neutrophil apoptosis. In addition, deposition of fragments of free light chains causes amyloidosis in several organs, including the liver, small intestine and heart, with subsequent end-organ injury, resulting in hepatomegaly, constipation and cardiomyopathy, respectively Citation[2,11,20–23,25,26].

Figure 2. A simplified diagram illustrating clinical manifestations and complications of multiple myeloma related to excess production of free light chains.Excess production of free light chains is responsible for tubular cast formation and obstruction (myeloma cast nephropathy), induction of proinflammatory cytokines and consequent direct tubular cell injury, functional impairment (Fanconi syndrome) and a state of chronic inflammation through interference with neutrophil apoptosis. In addition, deposition of fragments of free light chains causes amyloidosis in several organs, including the liver, small intestine and heart, with subsequent end-organ injury, resulting in hepatomegaly, constipation and cardiomyopathy, respectively Citation[2,11,20–23,25,26].
Figure 3. Flowchart illustrating the main pathogenetic molecular mechanisms involved within the renal parenchyma, with endocytosis of free light chains by proximal tubular cells as an important contributor, and bortezomib is shown targeting key steps essential for MM survival.

While the pathogenesis of MM is complex, bortezomib as a multifaceted proteosome inhibitor targets pivotal steps in the chain of pathogenetic steps involved in MM cell survival. It was recently discovered that the pathogenesis inside the renal parenchyma might be potentiated by the endocytosis of free light chains by proximal tubular cells. Key factors targeted by bortezomib include NF-κB, IL-6 and VEGF Citation[27,57–61].

BM: Bone marrow; BMEC: Bone marrow endothelial cell; MM: Multiple myeloma.

Figure 3. Flowchart illustrating the main pathogenetic molecular mechanisms involved within the renal parenchyma, with endocytosis of free light chains by proximal tubular cells as an important contributor, and bortezomib is shown targeting key steps essential for MM survival.While the pathogenesis of MM is complex, bortezomib as a multifaceted proteosome inhibitor targets pivotal steps in the chain of pathogenetic steps involved in MM cell survival. It was recently discovered that the pathogenesis inside the renal parenchyma might be potentiated by the endocytosis of free light chains by proximal tubular cells. Key factors targeted by bortezomib include NF-κB, IL-6 and VEGF Citation[27,57–61].BM: Bone marrow; BMEC: Bone marrow endothelial cell; MM: Multiple myeloma.
Figure 4. A simplified diagram illustrating the multiple myeloma cell interactions and the various mechanisms of action of thalidomide and bortezomib.

Multiple myeloma (MM) cells interact with surrounding osteoclasts, osteoblasts, endothelial cells and bone marrow (BM) stromal cells, which is crucial for their survival and growth. Immunomodulators and proteosome inhibitors used in current anti-MM regimens, including bortezomib and thalidomide, function by targeting those interactions and signaling pathways Citation[51–54,126–129]. Bortezomib’s mechanism of action on: (1) MM cell: bortezomib disrupts IL-6 and NF-κB; (2) Endothelial cell: bortezomib inhibits VEGF, angiogenesis and cell migration; (3) BM stromal cell: bortezomib reduces NF-κB-dependent secretion of cytokines; (4) Osteoblast: bortezomib-dependent elevation of Runx2 activity mediates osteoblast differentiation and so induces osteoblast activation Citation[55]. Thalidomide’s mechanism of action on: (A) MM cell: thalidomide causes MM cell growth arrest and apoptosis, as well as natural killer cell-mediated MM cell death via IL-2 and IFN-γ; (B) Osteoclast: thalidomide inhibits osteoclast-activating factors; (C) BM stromal cell: thalidomide inhibits adhesion of MM cells to BM stromal cells, inhibits IL-6 and TNF-α, and decreases VEGF and FGF, decreasing angiogenesis Citation[51–54,127–130].

Figure 4. A simplified diagram illustrating the multiple myeloma cell interactions and the various mechanisms of action of thalidomide and bortezomib.Multiple myeloma (MM) cells interact with surrounding osteoclasts, osteoblasts, endothelial cells and bone marrow (BM) stromal cells, which is crucial for their survival and growth. Immunomodulators and proteosome inhibitors used in current anti-MM regimens, including bortezomib and thalidomide, function by targeting those interactions and signaling pathways Citation[51–54,126–129]. Bortezomib’s mechanism of action on: (1) MM cell: bortezomib disrupts IL-6 and NF-κB; (2) Endothelial cell: bortezomib inhibits VEGF, angiogenesis and cell migration; (3) BM stromal cell: bortezomib reduces NF-κB-dependent secretion of cytokines; (4) Osteoblast: bortezomib-dependent elevation of Runx2 activity mediates osteoblast differentiation and so induces osteoblast activation Citation[55]. Thalidomide’s mechanism of action on: (A) MM cell: thalidomide causes MM cell growth arrest and apoptosis, as well as natural killer cell-mediated MM cell death via IL-2 and IFN-γ; (B) Osteoclast: thalidomide inhibits osteoclast-activating factors; (C) BM stromal cell: thalidomide inhibits adhesion of MM cells to BM stromal cells, inhibits IL-6 and TNF-α, and decreases VEGF and FGF, decreasing angiogenesis Citation[51–54,127–130].

Multiple myeloma (MM) is a clonal B-cell malignancy of the bone marrow (BM) associated with a variety of clinical manifestations, including hypercalcemia, renal impairment (RI), recurrent infection, anemia and bone disease Citation[1]. Reports indicate that MM patients with RI suffer more frequently from severe anemia, hypercalcemia, Bence–Jones proteinuria and skeletal abnormalities Citation[2]. The presence of such complications, and in particular RI, impacts adversely on quality of life, as well as survival, and is an important determinant of treatment choice Citation[3].

Although MM remains an incurable disease, the introduction of new drugs over the past decade has significantly improved patient outcome, extending the average life expectancy by between 2 and 3 years, from an average of 3–5 years to at least 5–7 years Citation[4,5].

RI represents a serious manifestation of progressive MM, and usually indicates substantial tumor burden and more aggressive disease, resulting in a higher early mortality, approaching 28% in some series Citation[6]. The pathophysiology of renal disease in MM is complex and is typically attributed to a variety of toxic effects on the kidney. This review includes an overview of MM and then focuses specifically on RI in MM, its causes and the manner in which it is best managed in the context of novel therapies.

Epidemiology

MM comprises approximately 1% of all malignancies and 13% of hematological malignancies Citation[7], with an annual incidence of approximately 20,000 cases per year in the USA Citation[8]. It is the second most common hematological malignancy after non-Hodgkin’s lymphoma Citation[9]. RI is present in approximately 20% of MM patients at the time of diagnosis (defined as a serum creatinine level of 177 µmol/l [2 mg/dl] or higher) and over 50% of patients at some stage in the course of their disease Citation[10,11]. Importantly, the risk of renal injury in MM is substantially increased in MM patients with light-chain paraproteinemias, especially λ light-chain disease Citation[2,3,10,12].

Clinical presentation

The presentation of MM can be insidious and is often detected in asymptomatic patients through routine blood testing, although symptoms are present in the majority of patients by the time of diagnosis. A retrospective study of 1027 patients with MM identified the most common presenting symptoms and signs, which included elevated serum creatinine (in 48% of patients), anemia (73%), bone pain (58%), fatigue (32%), hypercalcemia (13%), unexplained weight loss (24%), paresthesias (at least 5%), hepatomegaly (4%), splenomegaly (1%), lymphadenopathy (1%) and fever (0.7%) Citation[1]. In addition, extramedullary plasmacytomas are identified in at least 7% of MM patients at the time of diagnosis, and an increasing proportion of MM patients will develop extramedullary plasmacytomas later in the course of their disease Citation[13].

Patients with RI and monoclonal immunoglobulin (Ig) deposition disease often suffer from hypertension, while conversely those with amyloid deposition may suffer from hypotension Citation[3]. Symptoms suggestive of systemic amyloidosis or monoclonal Ig deposition include congestive heart failure, cardiac arrhythmias, hepatomegaly, portal hypertension and periorbital purpura Citation[3,14]. Furthermore, gastrointestinal bleeding and high serum alkaline phosphatase suggest amyloid deposition affecting the GI tract and liver, respectively Citation[3].

Diagnosis & evaluation

Diagnostic evaluation for patients with suspected MM includes obtaining the basic metabolic panel, serum β2-microglobulin level, complete blood count, serum and urine protein electrophoresis, serum free light chains (SFLCs) and assessment of end-organ function (specifically to assess for RI, anemia, hypercalcemia and bone disease) Citation[3,15]. Detection of serum or urine monoclonal Ig is critical in the diagnosis of MM Citation[3]. Importantly, those with RI secondary to Ig deposition with clonal plasma cells in their BM are identified as having symptomatic MM, regardless of other features, including the extent of the monoclonal protein or the percentage of BM plasma cells Citation[3,15]. Kidney biopsy may be considered in selected cases, including patients with significant nonselective proteinuria and predominant albuminuria, to assess for amyloidosis and monoclonal Ig deposition disease Citation[16].

BM aspiration and biopsy are performed to assess the extent of plasma cell involvement and evaluate for cytogenetic abnormalities. The evaluation of cytogenetic abnormalities includes both conventional metaphase chromosome analysis and FISH, targeting recurrent genetic aberrations such as deletion (del) 13, del 17, translocation (t) (4;14), t(11;14) and t(14;16) Citation[15]. Bone involvement is assessed using skeletal surveys, MRI and/or PET–computed tomography.

The SFLC assay is an important part of the diagnostic evaluation, particularly in patients with oligosecretory and nonsecretory disease. However, the use of SFLC assays in patients with RI can be challenging, as the clearance of all SFLCs decreases with decreasing glomerular filtration rate (GFR) Citation[17]. This leads to an increase in their half-life and an abnormal SFLC ratio, as reported in a recent study by Hutchison and colleagues Citation[17], which suggested that the reference range of SFLC ratio should be 0.37–3.1 (rather than the standard 0.26–1.65) in patients with RI requiring dialysis. Use of this modified range increases the specificity from 93 to 99% with no loss of sensitivity Citation[17]. Measurement of SFLC levels together with the κ/λ ratio with these extended reference ranges appears to aid the diagnosis of MM in patients with RI, and decreases the number of false-positive results, therefore allowing for rapid diagnosis and early initiation of treatment Citation[17]. Interestingly, it has also been noted that when cast nephropathy is the cause of RI, the SFLC levels tend to be much higher in comparison to other renal pathologies Citation[17], with other studies suggesting that high SFLC levels are associated with an elevated risk of renal injury in patients with newly diagnosed MM Citation[18]. Furthermore, recently developed nephelometric SFLC immunoassays have the ability to detect both monomers and dimers of κ and λ at concentrations less than 2–4 mg/l, without confounding the results from intact Ig, which may further refine the ability to evaluate tumor burden Citation[19].

Pathophysiologic mechanisms of renal injury in MM

The pathophysiology of RI in MM is complex and is associated with various underlying processes. Renal injury in the majority of cases is attributed to tubulointersitial damage, where ‘myeloma kidney’ results as a direct consequence of the high SFLC levels Citation[11]. Other causes include dehydration, hypercalcemia, hyperuricemia, amyloid deposition, infections, concomitant exposure to nephrotoxic medications, such as NSAIDs, plasma cell infiltration and contrast injury Citation[2,11].

SFLCs are primarily metabolized by the kidneys through glomerular filtration, followed by endocytosis in the proximal tubule and degradation within lysosomes Citation[20–22]. When Ig light chains are produced in excess, as in MM, absorption mechanisms are overwhelmed in the proximal tubule. Thus, light chains enter the distal tubules, with the subsequent appearance of Bence–Jones protein in urine. Bence–Jones protein then interacts with Tamm–Horsfall protein in the distal tubule, resulting in tubular cast formation and obstruction Citation[23]. This obstructive cast formation in the tubules with light chains and other MM proteins is classically referred to as myeloma kidney or ‘myeloma cast nephropathy’ (see ).

In addition, excess SFLC can cause direct injury to proximal tubular cells Citation[11], through the induction of proinflammatory cytokines (including IL-6, IL-8 and TNF) Citation[24] and other pathways leading to tubular cell death Citation[25]. They can also cause functional impairment, leading to Fanconi syndrome, which is characterized by proximal tubular damage in which glucose, amino acids, uric acid, phosphate and bicarbonate fail to be reabsorbed Citation[26]. Light chains also interfere with apoptosis of neutrophils, contributing to a state of chronic inflammation Citation[23]. Thus, urinary free light chains (FLCs) tend to cause renal injury through intratubular cast formation and direct tubular toxicity as well as inflammation (see ).

Most recently, it was found that endocytosis of light chains by proximal tubular cells might be a significant contributor to the chain of pathogenetic events, with subsequent activation of c-Src and NF-κB supporting MM cell survival (see ) Citation[27]. Moreover, deposition of monoclonal light chains can occur in several organs, including the kidney, heart, liver and small intestine, leading to the development of amyloid light chain (AL) amyloidosis or light chain deposition disease (LCDD) Citation[28]. LCDD is defined as the deposition of monoclonal amorphous light chains in multiple organs, which do not exhibit a fibrillar structure when examined ultrastructurally Citation[28]. LCDD pathogenesis is similar to that of AL amyloidosis, except that light chain fragments do not have the required biochemical characteristics to form amyloid fibrils Citation[28]. Notably, BM examination in patients with AL amyloidosis typically shows monoclonal plasma cells with overexpression of either κ or λ light chains with positive staining for CD138 and CD32B Citation[29].

Hypercalcemia usually results from enhanced bone resorption by activated osteoclasts, which may be further enhanced by the increased secretion of bone-resorbing cytokines, such as lymphotoxin and IL-6 Citation[30,31]. Hypercalcemia contributes to renal injury by causing vasoconstriction of the renal vessels, enhancing calcium deposition inside the renal tubules and augmenting the toxicity of filtered light chains Citation[32,33].

Hyperuricemia results from the accelerated catabolism of purine nucleic acids to hypoxanthine and xanthine and then to uric acid via the enzyme xanthine oxidase. Being poorly soluble in water, excess uric acid can in turn lead to renal injury via crystal precipitation and deposition in the renal tubules Citation[34,35].

Classification of renal disease with MM

Renal disease with MM can be classified either according to histological type or the primary site of injury (see Box 1). Several studies evaluating the histological characteristics of renal biopsy in MM patients with RI identified myeloma cast nephropathy as the major histological type, reaching as high as 60% of patients in some studies Citation[2,36]. Other histological types include tubulointerstitial nephritis, amyloidosis, acute tubular necrosis, nodular glomerulosclerosis and plasma cell infiltration (see ) Citation[2,36]. In addition, several studies have confirmed myeloma cast nephropathy as the predominant cause of dialysis-dependent RI in patients with MM Citation[37–39]. Similarly, nephrotic syndrome occurs in approximately 7% of individuals with RI Citation[2], and its presence usually suggests the presence of either amyloidosis Citation[40] or LCDD as the underlying etiology Citation[41]. Moreover, studies have identified precipitating factors of RI in MM patients as dehydration (19–33%), hypercalcemia (24–31%), nephrotoxic medications (15–16%), sepsis (9–23%), recent surgical procedures (5%) and contrast agents (2%) Citation[2,42]. Myeloma cast nephropathy represents the hallmark insult to the kidneys in the majority of patients.

Tumor burden & effect on prognosis

Early mortality in MM patients is typically attributable to the mutual effect of disease activity along with concurrent complications, including RI. A series of large multicenter trials assessing 3107 patients identified RI in 28% of patients who experienced early death (defined as death occurring within 60 days of diagnosis) Citation[6]. Early mortality could not otherwise be predicted using traditional prognostic characteristics, and thus it is recommended that all patients with RI be considered at high risk during induction therapy Citation[6].

Indeed, a recently published study of 198 patients with MM and RI has identified estimated (e)GFR <30 (or eGFR <50 in individuals >59 years of age) and β2-microglobulin as very important prognostic factors that affect overall survival (OS), and a novel MM risk-stratification score predicting survival was developed Citation[43]. Notably, the reversal of MM-associated RI is a key element of prognosis and often precedes the response to antimyeloma therapy as a predictor of improved survival Citation[3,44].

Management

Management of MM begins with general measures, including intravascular volume repletion, hemodynamic support, controlling hypercalcemia (using intravenous saline, steroids and bisphosphonate therapy, dose adjusted according to renal function) and treating hyperuricemia (using allopurinol, rasburicase and/or hemodialysis) Citation[45,46]. Certain medications should be avoided, including loop diuretics, NSAIDs and renin–angiotensin inhibitors. These standard management principles are often successful in correcting moderate degrees of RI Citation[3].

The treatment landscape for MM has changed substantially over the past decade with the introduction of bortezomib, lenalidomide and thalidomide. Combinations of these agents with dexamethasone are now considered as options for first-line therapy for induction, maintenance and salvage therapy Citation[47,48], with autologous stem cell transplantation (SCT) remaining a cornerstone of therapy for appropriately selected patients. Choice of treatment is guided by a number of factors, including age, comorbid conditions, eligibility for SCT and risk assessment, as determined by both International Staging System stage and cytogenetic findings Citation[49,50]. Recent accumulating evidence suggests that therapies with novel agents are changing the perception that RI per se is an ominous sign in the course of MM Citation[48]. We will review the use of these drugs both as single agents and in different combinations in this setting, and specifically in terms of efficacy, safety, potential dose adjustments and their impact on the reversibility of RI.

Bortezomib

Bortezomib is a 26S proteosome inhibitor that acts through various pathways (see ), including the disruption of IL-6 signaling Citation[51–54]. It enhances bone integrity by inducing osteogenic differentiation and through interactions with FGF2 and the PDZ-binding motif (TAZ), with increased expression of transcription factor Runx2 Citation[55]. It also protects renal tubular cells through its effect on the NF-κB pathway, leading to antiapoptotic gene expression Citation[56]. As illustrated in , MM pathogenesis within the renal parenchyma is contributed to by SFLC endocytosis through the proximal tubular cells, and bortezomib, as a multifaceted proteosome inhibitor, targets key factors involved, including NF-κB, IL-6 and VEGF Citation[27,57–61]. Interestingly, bortezomib also reduces the serum level of cystatin-C, a marker of RI Citation[62], especially in those with relapsed MM Citation[63]. Bortezomib is hepatically metabolized and its clearance is therefore independent of renal function. Dose adjustment is therefore not required in the setting of RI, and this together with its other properties makes it an ideal drug for use in patients with RI Citation[64].

The APEX Phase III study was the first large randomized controlled trial to show that bortezomib is more effective than high-dose dexamethasone in delaying disease progression in patients with relapsed MM Citation[65]. It included 669 patients with relapsed MM who were randomized to receive either bortezomib or high-dose dexamethasone, with results from this pivotal study leading to full US FDA approval for the use of bortezomib in the treatment of patients with relapsed disease. Patients treated with bortezomib achieved higher combined complete and partial response rates compared with high-dose dexamethasone (38 vs 18%; p < 0.001), significantly longer time to progression (6.22 vs 3.49 months; hazard ratio [HR]: 0.55; p < 0.001) and longer 1-year OS (80 vs 66%; p = 0.003) Citation[65]. Moreover, data from an updated analysis of the APEX study showed a median OS benefit of bortezomib over high-dose dexamethasone despite crossover (29.8 vs 23.7 months; p = 0.027) Citation[66], while results from the study also demonstrated that using bortezomib in those with RI was not associated with increased toxicity in comparison to those with normal renal function Citation[67,68]. In fact, rates of early discontinuation of treatment owing to serious side effects were higher with high-dose dexamethasone than with bortezomib Citation[67,68]. Response rate, time to progression, OS and safety were not affected by renal function, with OS significantly longer in those treated with bortezomib versus high-dose dexamethasone, suggesting that bortezomib was more effective in overcoming the detrimental effect of RI in MM Citation[68].

The benefit of bortezomib in MM patients with RI was also shown by a retrospective analysis involving 117 patients with MM and RI, including 14 who received dialysis. Overall, 41% of patients experienced reversal of RI after approximately 2.3 months of bortezomib treatment Citation[69], with three patients (21%) in the dialysis group coming off dialysis after 1–4 months. In terms of anti-MM effect, a partial response (PR) was observed in 73% of patients, complete response (CR) in 19% and near-CR in 8% Citation[69], with improvement in renal function seen among patients with different stages of RI and a 2-year OS estimated at approximately 50% Citation[69]. To assess bortezomib in the dialysis-dependent population, a retrospective analysis in 24 MM patients requiring dialysis showed that bortezomib as a single agent or in combination achieved an overall response rate of 75%, with 30% CR and near-CR. Notably, one patient was spared dialysis while three others became independent of dialysis Citation[70]. In another study of 20 patients with RI treated with bortezomib-based therapy, reversal of RI was evident in 40% of patients, with 85% of patients showing some decrease in serum creatinine Citation[71].

Similarly, bortezomib plus dexamethasone in 18 MM patients with RI achieved reversal of nephropathy in approximately 39% of patients, in addition to about 33% of patients showing 50% reduction in serum creatinine Citation[72]. Interestingly, all patients with a serum creatinine level <4 mg/dl normalized their renal function, in contrast to only one patient among those with serum creatinine >4 mg/dl, suggesting creatinine level as an independent prognostic factor of renal recovery in this setting Citation[72]. In terms of MM response, an overall response rate of approximately 83% was reported, including 50% CR, approximately 17% very good partial response (VGPR), with a correlation between MM response and recovery of renal function seen in 13 out of 15 patients Citation[72]. The use of bortezomib in other combinations, particularly the bortezomib–doxorubicin–dexamethasone regimen, has been shown to be highly effective in MM patients with light chain-induced RI Citation[73], with responses in 72% of patients overall, including CR or near-CR in 38% and VGPR in 15%, as well as improvement in renal function in 62% of patients Citation[73].

Finally, the use of bortezomib in newly diagnosed MM patients with RI was evaluated as part of the international Phase III VISTA study, comprising bortezomib, melphalan and prednisone (VMP) versus melphalan and prednisone (MP), with the results outlined in Citation[74].

Results overall showed that VMP is superior to MP in terms of response rates and renal reversibility, and outcomes were similar to those with normal renal function Citation[74]. Although adverse events (AEs) occurred more frequently with VMP in those with RI, the treatment duration, rates of drug discontinuation and dose reduction owing to AEs were the same in both individuals with RI and those with normal renal function. Peripheral neuropathy rates were not increased in those with impaired renal function. While it is not ideal to use melphalan in patients with severe RI because of the need for dose reduction and potentially increased toxicity, with other combinations excluding melphalan generally recommended, VMP proved to be superior to MP, with unaltered efficacy in patients with moderate RI Citation[74].

In summary, these results correlate with data from several other studies Citation[11,75–77] showing that bortezomib-based regimens are effective and safe in patients with RI, and lead to the reversal of RI in a significant number of cases, with bortezomib also having no detrimental effect on stem cell harvesting and engraftment in this setting Citation[78].

Lenalidomide

Lenalidomide is an immunomodulatory drug (IMiD) with pleiotropic anti-MM effects, including inhibition of proinflammatory cytokines, including TNF-α, downregulation of IL-6 and NF-κB, enhancement of natural killer cell activity, inhibition of angiogenesis and induction of apoptosis Citation[79–82]. Lenalidomide is predominantly excreted in urine (84% unchanged), and a single hemodialysis session can remove 31% of the dose Citation[83]. As RI progresses, lenalidomide’s clearance decreases to 69, 38 and 43% in those with mild (creatinine clearance [CrCl] >50 ml/min), moderate (CrCl <50 ml/min) and severe (CrCl <30 ml/min) RI, respectively Citation[83]. There is a 240 and 360% rise in the area under the curve of lenalidomide in patients with severe RI and end-stage renal failure, respectively Citation[83]. Therefore, lenalidomide dose adjustment is recommended for those with moderate and severe RI Citation[83].

The potent anti-MM activity of lenalidomide in conjunction with dexamethasone was demonstrated in two pivotal Phase III studies, where the combination proved superior to dexamethasone alone in terms of overall response (60–61% vs 19.9–24%; p < 0.001), CR (14.1–15.9% vs 0.6–3.4%; p < 0.001) time to progression (11.1–11.3 vs 4.7 months; p < 0.001), and OS (29.6 vs 20.2 months; p < 0.001) Citation[84,85]. Despite the fact that it is renally cleared, lenalidomide can be used in patients with RI (defined as CrCl <50 ml/min). This was demonstrated by a recent study involving 50 patients, >50% of whom were refractory to thalidomide and/or bortezomib, and 24% of whom had RI. The overall response rate was 58% in patients with RI, versus 60.5% in those with normal renal function Citation[86]. RI did not affect the response of MM to therapy, as reflected by a median progression-free survival of 9 months and OS of 16 months in all patients, regardless of renal function Citation[86]. As for the renal response in the RI group (16 patients), three (25%) achieved complete recovery, with two more (16%) showing some improvement. Importantly, when lenalidomide was dose-adjusted according to the CrCl, the incidence of AEs was also comparable in those with impaired renal function versus those with normal renal function Citation[86].

Given that earlier trials of lenalidomide excluded patients with serum creatinine levels >221 mol/l (creatinine >2.5 mg/dl) Citation[87], these results are useful, although it is important to note that certain toxicities associated with lenalidomide (especially myelosuppression) are more likely to occur if the drug is used in patients with RI Citation[88–90]. Furthermore, there have been isolated reports of progressive azotemia occurring during lenalidomide therapy in patients with MM and RI, so it is important to closely monitor renal function when lenalidomide is administered in this setting Citation[89]. In aggregate, lenalidomide is highly effective and can be used in MM with RI if necessary, with dose adjustment to avoid toxicity.

Thalidomide

Like lenalidomide, thalidomide is an IMiD and exerts its anti-tumor effect by acting on TNF-α Citation[91], natural killer cells, IL-2, IFN-γ, enhancement of cell-mediated immunity and modulating cellular adhesion molecules (see ) Citation[92–95]. Thalidomide undergoes non-enzymatic hydrolysis in plasma, forming multiple metabolites, with <1% urine excretion of unchanged drug Citation[96]. The exact metabolic route and fate of thalidomide is not yet understood Citation[96], and it does not appear to be hepatically metabolized to any large extent Citation[96].

Thalidomide, as a single agent or in combination, has proven to be useful in advanced and/or refractory MM, with a response rate ranging from 30 to 60% of patients Citation[97–100]. In a study of 20 patients with RI (defined as serum creatinine >130 mmol/l) and advanced relapsed/refractory MM, in which three patients requiring chronic hemodialysis were included, the efficacy of thalidomide was evaluated. Eight patients received thalidomide as a single agent, while 12 patients received thalidomide plus dexamethasone. A >50% decrease in serum or urine M-component was observed in nine patients (45%), seven of whom were treated with thalidomide and dexamethasone and two with thalidomide alone. Six additional patients achieved a minimal response (defined as >25% paraprotein decrease). The overall response rate was reported as 75%, while improvement of renal function was observed in 12 out of 15 responding patients Citation[50]. Two patients who were dependent on chronic hemodialysis showed a decrease in serum creatinine, and the side effects of thalidomide as a single agent or in combination did not appear to be affected by RI Citation[50]. Studies suggest that serum levels of thalidomide are not affected by either RI or by hemodialysis Citation[101]. It is thus considered safe to administer thalidomide in patients with advanced MM and RI without requiring dose modification, although in general dose reduction is considered prudent Citation[50,101].

Treating patients with MM and associated light chain-associated amyloidosis resulting in RI is challenging, and thalidomide has also been assessed in this patient population. In a study of 16 patients with MM and amyloidosis, of whom 14 had RI, a response was seen in 25% of patients Citation[102], which is similar to patients with other types of MM. However, AEs were frequent, as reflected by 25% of patients requiring dose reduction and another 25% discontinuing treatment altogether Citation[102].

Recently, the use of thalidomide plus dexamethasone as induction therapy prior to autologous SCT was reported in 31 patients with MM and RI Citation[103], resulting in PR or better of 74%, including 26% VGPR, which are comparable to those seen in patients with normal renal function in this setting Citation[104–106]. Interestingly, improvement in renal function occurred in 82% of those achieving PR, versus 37% in those who failed to achieve PR. Peripheral blood stem cell mobilization was successful in 26 patients, and SCT was subsequently performed in 22 patients, suggesting that the combination of thalidomide and dexamethasone can be effective when used in conjunction with SCT in patients with MM and RI Citation[103].

Importantly, the use of lower-dose thalidomide in patients with relapsed or refractory MM has been described with comparable response rates and lower toxicity, with no benefit of dose escalation seen Citation[107]. In summary, thalidomide is relatively safe and effective to use in MM patients with RI; however, dose reduction is recommended in patients with light chain-associated amyloidosis in particular Citation[108].

Reversibility of RI in the era of newer agents

Several studies investigating bortezomib, lenalidomide and thalidomide have shown impressive activity in reversing renal dysfunction (see ) over and above their well-established anti-myeloma effects Citation[11]. For example, in a study of 41 MM patients receiving high-dose dexamethasone with or without thalidomide and/or bortezomib, reversal of RI was shown in 73% of patients when dexamethasone was used alone (median duration of 1.9 months) and in 80% when it was used with thalidomide and/or bortezomib (median duration of 0.8 months) Citation[109]. Predictors of failure to reverse renal function included more severe RI and marked Bence–Jones proteinuria prior to therapy. Consistent with other trials, in patients whose MM responded to treatment, the reversal of RI was 85%, whereas in patients not significantly responding, reversal of RI was less at 56% Citation[109].

Similarly, bortezomib plus dexamethasone in 46 patients with RI and MM showed a renal response in 59% of patients, including 30% complete renal response, with two out of nine patients no longer requiring dialysis Citation[110], with a similar safety profile to those with normal renal function. Previously untreated patients had a greater chance of complete renal recovery and those with light chain MM also had a higher chance of achieving renal response in a shorter time frame. Interestingly, in a subset of patients, cystatin-C levels >2 mg/l or those with a cystatin-C-calculated eGFR <30 ml/min were associated with lower chances of complete renal response Citation[110].

In a recent study evaluating the use of dexamethasone and/or thalidomide in 12 patients requiring dialysis, encouraging results have also been reported Citation[111]. Complete response and partial response were achieved in three and five patients, respectively. Eight cases (67%) became dialysis independent, including six treated with high-dose dexamethasone and two treated with a thalidomide-based regimen, after a median duration of 2 months Citation[111]. It was noted that a high SFLC level correlated with the presence of advanced RI, and a significant drop in SFLC level preceded improvement of renal function and dialysis independence. It was concluded that dialysis-dependent RI can be reversed by dexamethasone and/or thalidomide even in advanced age, and that SFLC levels could be used in predicting the improvement and reversibility of renal function Citation[111].

Finally, the use of different novel regimens as frontline therapies for reversing RI in MM patients has been evaluated in a large trial that included 96 consecutive patients with RI (defined as CrCl <50 ml/min), where 32 patients received conventional chemotherapy plus dexamethasone (VAD, VAD-like regimens or melphalan plus dexamethasone), 47 received IMiD-based regimens (thalidomide or lenalidomide with high-dose dexamethasone and/or cyclophosphamide or melphalan), and 17 patients received bortezomib- and dexamethasone-based regimens Citation[112]. This study showed improvement of RI in 79 and 94% of patiens in the IMiD and bortezomib-treated groups, respectively, compared with 59% in the conventional chemotherapy-treated group (p = 0.02) Citation[112]. Bortezomib therapy and CrCl >30 ml/min were identified as independent factors associated with a higher probability of achieving renal response and a shorter median time to achieve renal recovery Citation[112]. The authors concluded that bortezomib-based regimens are preferred as frontline therapy for newly diagnosed myeloma patients with RI, which is a guideline now generally accepted in the field, on the basis of these results and other studies summarized previously Citation[11,67–78].

Other treatment options

HCO: a new hope?

Based on the understanding that tubulointerstitial injury in MM occurs as a direct outcome of elevated SFLC levels, recent studies have investigated the benefit of direct SFLC removal on renal recovery. Although traditional plasma exchange is estimated to remove approximately 25% of the total SFLCs, a randomized clinical trial showed no significant clinical benefit of its use for preserving renal function Citation[113]. One explanation might be that traditional plasma exchange only removes intravascular SFLCs with no effect on the extravascular SFLCs, which soon redistribute into the intravascular compartment. However, an additional promise has emerged with the development of high cut-off hemodialysis (HCO), as extended therapy allows the removal of both intravascular SFLCs and extravascular FLCs, with model calculations suggesting that 90% of FLCs can be removed by utilizing HCO in a 3-week period Citation[114]. In this study, HCO succeeded in decreasing SFLCs by 35–70% in 2 h, although SFLC concentrations showed some rebound on subsequent days unless coupled with effective chemotherapy Citation[114].

Faced with these encouraging results, a study was conducted investigating the clinical benefit of combining standard chemotherapy with HCO. In a total of 19 patients with acute renal failure secondary to MM, this trial showed that 13 patients (68%) succeeded in maintaining reduced SFLC levels (median 85%), with recovery of renal function to become dialysis independent (median duration 27 days). Patients achieving renal function recovery had significantly improved survival (p < 0.012) Citation[115]. Another study of 39 patients with myeloma kidney showed that lowered SFLC levels significantly predicted recovery of renal functions (p = 0.003), demonstrating by day 21 a decrease of SFLC levels by 60% was associated with renal recovery in 80% of the patients. Strikingly, it showed that median survival was 42.7 months for patients with renal recovery, in contrast to 7.8 months for patients who did not recover renal function (p < 0.02), again supporting the notion that RI is a strong factor affecting survival Citation[116]. With such data suggesting HCO as a hopeful new modality in the management of MM and RI, it will be important that future large-scale studies are conducted to confirm its benefit.

Integrating therapeutic modalities

Dialysis as part of renal replacement therapy is key for patients whose renal function fails to rapidly reverse with combination therapy utilizing bortezomib, dexamethasone and IMiDs, with or without the addition of cyclophosphamide Citation[49]. Kidney transplantation is also an alternative treatment option to dialysis, although with limited success owing to the risk of MM recurrence and infections Citation[3]. Integrating both renal transplantation and SCT in younger patients with HLA-identical siblings is also under study Citation[3].

The use of high-dose melphalan in RI patients has proven to be toxic, so if melphalan is to be used the dose should typically not exceed 140 mg/m2, to avoid serious AEs Citation[117]. For patients with persistent RI, combination chemotherapy (e.g., doxorubicin in combination with bortezomib and dexamethasone) is another option Citation[117]. SCT remains a reasonable treatment, especially for resistant/relapsed cases of MM with moderate RI (serum creatinine >2 mg/dl), with successful induction followed by stem cell mobilization using cyclophosphamide 2.5–3 g/m2 plus melphalan 140–150 mg/m2, followed by autologous SCT as one possible sequence Citation[49].

With data suggesting a primary role of the proximal tubular cells in the pathogenesis of myeloma kidney (see ), newer agents are being developed, such as PACAP-38, which has been shown to prevent the process of proximal tubular cell activation by the FLCs, as well as inhibition of TNF-α and IL-6 Citation[118]. These agents remain experimental; however, they may have promise as a new generation of immune modulators with the potential to augment existing treatment strategies.

Expert commentary

RI is present in approximately 50% of MM patients at some point in the course of their disease. As it is the most common cause of early mortality in MM, patients with RI during initial therapy should be considered high risk, especially during induction, with myeloma cast nephropathy the most common cause of serious renal compromise. Diagnosis and treatment at the early stages, while renal injury is still reversible, is thus fundamental for improving prognosis Citation[117]. Diagnosis depends on traditional investigation, with special consideration to SFLC levels and SFLC ratios, while also considering the suggested extended reference ranges described previously.

Treatment with bortezomib, lenalidomide and thalidomide in patients with RI has proven to be of great value. Bortezomib has been shown to be safe and effective, and can lead to reversal of RI in a considerable number of patients, with evidence that bortezomib is a cornerstone drug in current combination therapies. In addition, there is sufficient evidence to confirm bortezomib’s safety in patients with RI, with the hope of renal recovery in a substantial portion of patients. Lenalidomide is renally cleared and thus severe RI can be associated with elevated serum levels and potentially higher toxicity. However, its use is often essential, so in patients with severe RI, dose adjustment is necessary to avoid toxicity, most notably myelosuppression. Thalidomide is also useful for salvage of patients with advanced MM, and all three agents have been used successfully in achieving reversal of RI. Melphalan can be toxic in patients with RI, and high-dose therapy should be used cautiously with dose reduction, as clinically indicated. Cyclophosphamide as an alternate alkylator is potentially more efficacious in this setting, and can be safely used, especially in combination.

Five-year view

The management of MM is constantly evolving as newer agents are developed. Although substantial data emphasize the overall benefit of novel agents in MM therapy, additional trials are required to identify which combination regimen gives the best long-term survival in this group of patients. We believe that novel therapies will continue to stand the test of time while evolving into newer generations, with particular emphasis on bortezomib and lenalidomide as platform drugs. Newer generations of current novel agents might aim on increasing efficacy, reducing AEs, and improving safety, especially in patients with RI. With recent data suggesting pomalidomide as one of the most potent immune-modulators Citation[119,120], we believe future studies investigating its safety and efficacy in patients with RI are critical.

SCT still constitutes a cornerstone of therapy in conjunction with novel agents in younger patients; however, its optional use as a treatment approach in RI remains debatable. Further clinical trials are needed to identify which line of therapy confers the best overall long-term survival.

Newer modalities include extended HCO, which has shown promising results, but still needs further large-scale studies to support its benefit. Moreover, newer agents, such as PACAP-38, offer the hope of future additions to our current arsenal of novel agents.

Traditionally, RI has been known to be an adverse prognostic and survival factor. New treatments and novel combinations may be changing this as the treatment paradigm in MM continues to evolve Citation[121]. Use of these newer agents upfront in patients with RI is recommended, provided dose reduction is used when indicated. Future large-scale studies are necessary to compare the efficacy and safety of these newer drugs in different novel combinations to be used in patients with MM and RI, and beyond. Moreover, these should provide sufficient data to formulate future management strategies involving newer drugs, as well as defining a clearer role for SCT in MM patients with RI and thus further improve patient outcome. Meanwhile, the search for newer agents will continue, with today’s innovation at both the bench and the bedside providing real hope for tomorrow Citation[122].

Table 1. Tabulated data reflecting the frequency of different histologic types of renal injury found in multiple myeloma patients with renal impairment according to Herrera et al. and Sakhuja et al.

Table 2. Renal impairment reversal rates according to the Phase III VISTA study.

Table 3. Summary of renal response in multiple myeloma patients treated with various newer anti-myeloma regimens.

Box 1. Classification of kidney disease in multiple myeloma according to the primary site of injury.

Glomerular

  • • Primary amyloidosis (AL or AH)

  • • Monoclonal immunoglobulin deposition:

    • – Light chain deposition disease

      – Heavy chain deposition disease

      – Light and heavy chain deposition disease

  • • Miscellaneous (cryoglobulinemia, proliferative glomerulonephritis)

Tubular

  • • Myeloma kidney (light chain cast nephropathy)

  • • Distal tubular dysfunction

  • • Proximal tubule dysfunction or acquired Fanconi syndrome

Interstitial

  • • Plasma cell infiltration

  • • Interstitial nephritis

Other causes

  • • Hyperuricemia

  • • Hypercalcemia

  • • Drugs (e.g., NSAIDs)

Key issues

  • • Renal impairment (RI) is a major problem complicating multiple myeloma (MM), being present in approximately 50% of patients during the course of their illness, with myeloma cast nephropathy representing the most common cause.

  • • Newer agents, including bortezomib, lenalidomide and dexamethasone, have revolutionized MM management, and as part of their anti-MM effect, can help improve renal function.

  • • Bortezomib-based regimens are effective and safe in patients with RI, and lead to the reversal of RI in a significant number of patients.

  • • For newly diagnosed MM patients with RI, bortezomib-based regimens are preferred as the first-line therapy.

  • • Since bortezomib is hepatically metabolized, its clearance is independent of renal function, and thus dose adjustment is not required in the setting of RI.

  • • Since it is renally excreted, lenalidomide use in moderate and severe RI requires dose adjustment. Studies show that RI did not impact response to therapy, and a significant proportion of patients showed renal improvement.

  • • Concerns about using lenalidomide with RI include myelosuppression and progressive azotemia, necessitating close monitoring of complete blood count and renal function.

  • • The combination of thalidomide and dexamethasone can be effective when used in conjunction with stem cell transplantation in patients with MM and RI.

  • • Thalidomide is relatively safe and effective to use in MM patients with RI; however, dose reduction is usually recommended in patients with light chain-associated amyloidosis, in particular.

  • • High cut-off hemodialysis (HCO) with improved light chain removal has promise as a supportive modality.

  • • Melphalan can be toxic in patients with RI, and high-dose therapy should be used cautiously with dose reduction as clinically indicated.

  • • Future studies are necessary to evaluate different combinations of novel agents, while defining clearer indications for stem cell transplantation among the therapeutic algorithm.

Acknowledgements

The authors gratefully acknowledge the assistance of Michelle Maglio with this manuscript.

References

  • Kyle RA, Gertz MA, Witzig TE et al. Review of 1027 patients with newly diagnosed multiple myeloma. Mayo Clin. Proc.78(1), 21–33 (2003).
  • Sakhuja V, Jha V, Varma S et al. Renal involvement in multiple myeloma: a 10-year study. Ren. Fail.22(4), 465–477 (2000).
  • Heher EC, Goes NB, Spitzer TR et al. Kidney disease associated with plasma cell dyscrasias. Blood116(9), 1397–1404 (2010).
  • Kumar SK, Rajkumar SV, Dispenzieri A et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood111(5), 2516–2520 (2008).
  • Munshi NC. Plasma cell disorders: an historical perspective. Hematology Am. Soc. Hematol. Educ. Program2008(1), 297 (2008).
  • Augustson BM, Begum G, Dunn JA et al. Early mortality after diagnosis of multiple myeloma: analysis of patients entered onto the United Kingdom medical research council trials between 1980 and 2002 – Medical Research Council Adult Leukaemia Working Party. J. Clin. Oncol.23(36), 9219–9226 (2005).
  • Raab MS, Podar K, Breitkreutz I, Richardson PG, Anderson KC. Multiple myeloma. Lancet374(9686), 324–339 (2009).
  • Coleman EA, Lynch H, Enderlin C et al. Initial report of a family registry of multiple myeloma. Cancer Nurs.32(6), 456–464 (2009).
  • Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J. Clin.59(4), 225–249 (2009).
  • Knudsen LM, Hippe E, Hjorth M, Holmberg E, Westin J. Renal function in newly diagnosed multiple myeloma – a demographic study of 1353 patients. The Nordic Myeloma Study Group. Eur. J. Haematol.53(4), 207–212 (1994).
  • Dimopoulos MA, Kastritis E, Rosinol L, Blade J, Ludwig H. Pathogenesis and treatment of renal failure in multiple myeloma. Leukemia22(8), 1485–1493 (2008).
  • Greipp PR, San Miguel J, Durie BG et al. International staging system for multiple myeloma. J. Clin. Oncol.23(15), 3412–3420 (2005).
  • Annesley TM, Burritt MF, Kyle RA. Artifactual hypercalcemia in multiple myeloma. Mayo Clin. Proc.57(9), 572–575 (1982).
  • Pozzi C, D’amico M, Fogazzi GB et al. Light chain deposition disease with renal involvement: clinical characteristics and prognostic factors. Am. J. Kidney Dis.42(6), 1154–1163 (2003).
  • Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report of the International Myeloma Working Group. Br. J. Haematol.121(5), 749–757 (2003).
  • Dimopoulos MA, Terpos E, Chanan-Khan A et al. Renal impairment in patients with multiple myeloma: a consensus statement on behalf of the International Myeloma Working Group. J. Clin. Oncol.28(33), 4976–4984 (2010).
  • Hutchison CA, Plant T, Drayson M et al. Serum free light chain measurement aids the diagnosis of myeloma in patients with severe renal failure. BMC Nephrol.9, 11 (2008).
  • Van Rhee F, Bolejack V, Hollmig K et al. High serum-free light chain levels and their rapid reduction in response to therapy define an aggressive multiple myeloma subtype with poor prognosis. Blood110(3), 827–832 (2007).
  • Bradwell AR, Carr-Smith HD, Mead GP et al. Highly sensitive, automated immunoassay for immunoglobulin free light chains in serum and urine. Clin. Chem.47(4), 673–680 (2001).
  • Santostefano M, Zanchelli F, Zaccaria A, Poletti G, Fusaroli M. The ultrastructural basis of renal pathology in monoclonal gammopathies. J. Nephrol.18(6), 659–675 (2005).
  • Batuman V, Guan S. Receptor-mediated endocytosis of immunoglobulin light chains by renal proximal tubule cells. Am. J. Physiol.272(4 Pt 2), F521–F530 (1997).
  • Batuman V, Verroust PJ, Navar GL et al. Myeloma light chains are ligands for cubilin (gp280). Am. J. Physiol.275(2 Pt 2), F246–F254 (1998).
  • Cohen G, Horl WH. Free immunoglobulin light chains as a risk factor in renal and extrarenal complications. Semin. Dial.22(4), 369–372 (2009).
  • Sengul S, Zwizinski C, Simon EE, Kapasi A, Singhal PC, Batuman V. Endocytosis of light chains induces cytokines through activation of NF-kappaB in human proximal tubule cells. Kidney Int.62(6), 1977–1988 (2002).
  • Sanders PW, Herrera GA, Galla JH. Human Bence Jones protein toxicity in rat proximal tubule epithelium in vivo. Kidney Int.32(6), 851–861 (1987).
  • Ma CX, Lacy MQ, Rompala JF et al. Acquired Fanconi syndrome is an indolent disorder in the absence of overt multiple myeloma. Blood104(1), 40–42 (2004).
  • Ying WZ, Wang PX, Aaron KJ, Basnayake K, Sanders PW. Immunoglobulin light chains activate nuclear factor-kappaB in renal epithelial cells through a Src-dependent mechanism. Blood117(4), 1301–1307 (2011).
  • Buxbaum JN, Chuba JV, Hellman GC, Solomon A, Gallo GR. Monoclonal immunoglobulin deposition disease: light chain and light and heavy chain deposition diseases and their relation to light chain amyloidosis. Clinical features, immunopathology, and molecular analysis. Ann. Intern. Med.112(6), 455–464 (1990).
  • Zhou P, Comenzo RL, Olshen AB et al. CD32B is highly expressed on clonal plasma cells from patients with systemic light-chain amyloidosis and provides a target for monoclonal antibody-based therapy. Blood111(7), 3403–3406 (2008).
  • Teoh G, Anderson KC. Interaction of tumor and host cells with adhesion and extracellular matrix molecules in the development of multiple myeloma. Hematol. Oncol. Clin. North Am.11(1), 27–42 (1997).
  • Cheung WC, Van Ness B. Distinct IL-6 signal transduction leads to growth arrest and death in B cells or growth promotion and cell survival in myeloma cells. Leukemia16(6), 1182–1188 (2002).
  • Smolens P, Barnes JL, Kreisberg R. Hypercalcemia can potentiate the nephrotoxicity of Bence Jones proteins. J. Lab. Clin. Med.110(4), 460–465 (1987).
  • Benabe JE, Martinez-Maldonado M. Hypercalcemic nephropathy. Arch. Intern. Med.138(5), 777–779 (1978).
  • Kjellstrand CM, Cambell DC 2nd, Von Hartitzsch B, Buselmeier TJ. Hyperuricemic acute renal failure. Arch. Intern. Med.133(3), 349–359 (1974).
  • Rieselbach RE, Bentzel CJ, Cotlove E, Frei E 3rd, Freireich EJ. Uric acid excretion and renal function in the acute hyperuricemia of leukemia. Pathogenesis and therapy of uric acid nephropathy. Am. J. Med.37, 872–883 (1964).
  • Herrera GA, Joseph L, Gu X, Hough A, Barlogie B. Renal pathologic spectrum in an autopsy series of patients with plasma cell dyscrasia. Arch. Pathol. Lab. Med.128(8), 875–879 (2004).
  • Irish AB, Winearls CG, Littlewood T. Presentation and survival of patients with severe renal failure and myeloma. QJM90(12), 773–780 (1997).
  • Johnson WJ, Kyle RA, Pineda AA, O’Brien PC, Holley KE. Treatment of renal failure associated with multiple myeloma. Plasmapheresis, hemodialysis, and chemotherapy. Arch. Intern. Med.150(4), 863–869 (1990).
  • Pozzi C, Pasquali S, Donini U et al. Prognostic factors and effectiveness of treatment in acute renal failure due to multiple myeloma: a review of 50 cases. Report of the Italian Renal Immunopathology Group. Clin. Nephrol.28(1), 1–9 (1987).
  • Kyle RA, Greipp PR. Amyloidosis (AL). Clinical and laboratory features in 229 cases. Mayo Clin. Proc.58(10), 665–683 (1983).
  • Randall RE, Williamson WC Jr, Mullinax F, Tung MY, Still WJ. Manifestations of systemic light chain deposition. Am. J. Med.60(2), 293–299 (1976).
  • Prakash J, Niwas SS, Parekh A et al. Multiple myeloma – presenting as acute kidney injury. J. Assoc. Physicians India57, 23–26 (2009).
  • Kleber M, Ihorst G, Deschler B et al. Detection of renal impairment as one specific comorbidity factor in multiple myeloma: multicenter study in 198 consecutive patients. Eur. J. Haematol.83(6), 519–527 (2009).
  • Knudsen LM, Hjorth M, Hippe E. Renal failure in multiple myeloma: reversibility and impact on the prognosis. Nordic Myeloma Study Group. Eur. J. Haematol.65(3), 175–181 (2000).
  • Niesvizky R, Badros AZ. Complications of multiple myeloma therapy, part 2: risk reduction and management of venous thromboembolism, osteonecrosis of the jaw, renal complications, and anemia. J. Natl Compr. Canc. Netw.8(Suppl. 1), S13–S20 (2010).
  • Gleason C, Nooka A, Lonial S. Supportive therapies in multiple myeloma. J. Natl Compr. Canc. Netw.7(9), 971–979 (2009).
  • Minnema MC, Van Der Spek E, Van De Donk NW, Lokhorst HM. New developments in the treatment of patients with multiple myeloma. Neth. J. Med.68(1), 24–32 (2010).
  • Ludwig H, Beksac M, Blade J et al. Current multiple myeloma treatment strategies with novel agents: a European perspective. Oncologist15(1), 6–25 (2010).
  • Pandit SR, Vesole DH. Management of renal dysfunction in multiple myeloma. Curr. Treat. Options Oncol.4(3), 239–246 (2003).
  • Tosi P, Zamagni E, Cellini C et al. Thalidomide alone or in combination with dexamethasone in patients with advanced, relapsed or refractory multiple myeloma and renal failure. Eur. J. Haematol.73(2), 98–103 (2004).
  • Adams J. The development of proteasome inhibitors as anticancer drugs. Cancer Cell5(5), 417–421 (2004).
  • Joazeiro CA, Anderson KC, Hunter T. Proteasome inhibitor drugs on the rise. Cancer Res.66(16), 7840–7842 (2006).
  • Hideshima T, Chauhan D, Hayashi T et al. Proteasome inhibitor PS-341 abrogates IL-6 triggered signaling cascades via caspase-dependent downregulation of gp130 in multiple myeloma. Oncogene22(52), 8386–8393 (2003).
  • Richardson PG, Hideshima T, Mitsiades C, Anderson K. Proteasome inhibition in hematologic malignancies. Ann. Med.36(4), 304–314 (2004).
  • Eda H, Aoki K, Kato S et al. The proteasome inhibitor bortezomib inhibits FGF-2-induced reduction of TAZ levels in osteoblast-like cells. Eur. J. Haematol.85(1), 68–75 (2010).
  • Sarkozi R, Perco P, Hochegger K et al. Bortezomib-induced survival signals and genes in human proximal tubular cells. J. Pharmacol. Exp. Ther.327(3), 645–656 (2008).
  • Hideshima T, Ikeda H, Chauhan D et al. Bortezomib induces canonical nuclear factor-kappaB activation in multiple myeloma cells. Blood114(5), 1046–1052 (2009).
  • Roccaro AM, Hideshima T, Raje N et al. Bortezomib mediates antiangiogenesis in multiple myeloma via direct and indirect effects on endothelial cells. Cancer Res.66(1), 184–191 (2006).
  • Petrucci MT, Ricciardi MR, Gregorj C et al. Effects of IL-6 variants in multiple myeloma: growth inhibition and induction of apoptosis in primary cells. Leuk. Lymphoma43(12), 2369–2375 (2002).
  • Kumar S, Witzig TE, Timm M et al. Expression of VEGF and its receptors by myeloma cells. Leukemia17(10), 2025–2031 (2003).
  • Vanderkerken K, Vande Broek I, Eizirik DL et al. Monocyte chemoattractant protein-1 (MCP-1), secreted by bone marrow endothelial cells, induces chemoattraction of 5T multiple myeloma cells. Clin. Exp. Metastasis19(1), 87–90 (2002).
  • Hojs R, Bevc S, Ekart R, Gorenjak M, Puklavec L. Serum cystatin C as an endogenous marker of renal function in patients with mild to moderate impairment of kidney function. Nephrol. Dial. Transplant.21(7), 1855–1862 (2006).
  • Terpos E, Katodritou E, Tsiftsakis E et al. Cystatin-C is an independent prognostic factor for survival in multiple myeloma and is reduced by bortezomib administration. Haematologica94(3), 372–379 (2009).
  • Mulkerin D, Remick S, Ramanathan R et al. A dose-escalating and pharmacologic study of bortezomib in adult cancer patients with impaired renal function. J. Clin. Oncol.24(18 Suppl.), 2032 (2006).
  • Richardson PG, Sonneveld P, Schuster MW et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N. Engl. J. Med.352(24), 2487–2498 (2005).
  • Richardson PG, Sonneveld P, Schuster M et al. Extended follow-up of a Phase 3 trial in relapsed multiple myeloma: final time-to-event results of the APEX trial. Blood110(10), 3557–3560 (2007).
  • Jagannath S, Barlogie B, Berenson JR et al. Bortezomib in recurrent and/or refractory multiple myeloma. Initial clinical experience in patients with impared renal function. Cancer103(6), 1195–1200 (2005).
  • San-Miguel JF, Richardson PG, Sonneveld P et al. Efficacy and safety of bortezomib in patients with renal impairment: results from the APEX Phase 3 study. Leukemia22(4), 842–849 (2008).
  • Morabito F, Gentile M, Ciolli S et al. Safety and efficacy of bortezomib-based regimens for multiple myeloma patients with renal impairment: a retrospective study of Italian Myeloma Network GIMEMA. Eur. J. Haematol.84(3), 223–228 (2010).
  • Chanan-Khan AA, Kaufman JL, Mehta J et al. Activity and safety of bortezomib in multiple myeloma patients with advanced renal failure: a multicenter retrospective study. Blood109(6), 2604–2606 (2007).
  • Roussou M, Kastritis E, Migkou M et al. Treatment of patients with multiple myeloma complicated by renal failure with bortezomib-based regimens. Leuk. Lymphoma49(5), 890–895 (2008).
  • Li J, Zhou DB, Jiao L et al. Bortezomib and dexamethasone therapy for newly diagnosed patients with multiple myeloma complicated by renal impairment. Clin. Lymphoma Myeloma9(5), 394–398 (2009).
  • Ludwig H, Adam Z, Hajek R et al. Light chain-induced acute renal failure can be reversed by bortezomib-doxorubicin-dexamethasone in multiple myeloma: results of a Phase II study. J. Clin. Oncol.28(30), 4635–4641 (2010).
  • Dimopoulos MA, Richardson PG, Schlag R et al. VMP (bortezomib, melphalan, and prednisone) is active and well tolerated in newly diagnosed patients with multiple myeloma with moderately impaired renal function, and results in reversal of renal impairment: cohort analysis of the Phase III VISTA study. J. Clin. Oncol.27(36), 6086–6093 (2009).
  • Ludwig H, Drach J, Graf H, Lang A, Meran JG. Reversal of acute renal failure by bortezomib-based chemotherapy in patients with multiple myeloma. Haematologica92(10), 1411–1414 (2007).
  • Malani AK, Gupta V, Rangineni R. Bortezomib and dexamethasone in previously untreated multiple myeloma associated with renal failure and reversal of renal failure. Acta Haematol.116(4), 255–258 (2006).
  • Qayum A, Aleem A, Al Diab AR, Niaz F, Al Momen AK. Rapid improvement in renal function in patients with multiple myeloma and renal failure treated with bortezomib. Saudi J. Kidney Dis. Transpl.21(1), 63–68 (2010).
  • Oakervee H, Popat R, Cavenagh JD. Use of bortezomib as induction therapy prior to stem cell transplantation in frontline treatment of multiple myeloma: impact on stem cell harvesting and engraftment. Leuk. Lymphoma48(10), 1910–1921 (2007).
  • Knight R. IMiDs: a novel class of immunomodulators. Semin. Oncol.32(4 Suppl. 5), S24–S30 (2005).
  • Davies FE, Raje N, Hideshima T et al. Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood98(1), 210–216 (2001).
  • Hayashi T, Hideshima T, Akiyama M et al. Molecular mechanisms whereby immunomodulatory drugs activate natural killer cells: clinical application. Br. J. Haematol.128(2), 192–203 (2005).
  • Richardson PG, Schlossman RL, Weller E et al. Immunomodulatory drug CC-5013 overcomes drug resistance and is well tolerated in patients with relapsed multiple myeloma. Blood100(9), 3063–3067 (2002).
  • Chen N, Lau H, Kong L et al. Pharmacokinetics of lenalidomide in subjects with various degrees of renal impairment and in subjects on hemodialysis. J. Clin. Pharmacol.47(12), 1466–1475 (2007).
  • Weber DM, Chen C, Niesvizky R et al. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N. Engl. J. Med.357(21), 2133–2142 (2007).
  • Dimopoulos M, Spencer A, Attal M et al. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N. Engl. J. Med.357(21), 2123–2132 (2007).
  • Dimopoulos MA, Christoulas D, Roussou M et al. Lenalidomide and dexamethasone for the treatment of refractory/relapsed multiple myeloma: dosing of lenalidomide according to renal function and effect on renal impairment. Eur. J. Haematol.85(1), 1–5 (2010).
  • Weber D, Wang M, Chen C et al. Lenalidomide plus high-dose dexamethasone provides improved overall survival compared to high-dose dexamethasone alone for relapsed or refractory multiple myeloma (MM): results of 2 Phase III studies (MM-009, MM-010) and subgroup analysis of patients with impaired renal function. ASH Annual Meeting Abstracts108(11), 3547 (2006).
  • Wang M, Dimopoulos MA, Chen C et al. Lenalidomide plus dexamethasone is more effective than dexamethasone alone in patients with relapsed or refractory multiple myeloma regardless of prior thalidomide exposure. Blood112(12), 4445–4451 (2008).
  • Batts ED, Sanchorawala V, Hegerfeldt Y, Lazarus HM. Azotemia associated with use of lenalidomide in plasma cell dyscrasias. Leuk. Lymphoma49(6), 1108–1115 (2008).
  • Niesvizky R, Naib T, Christos PJ et al. Lenalidomide-induced myelosuppression is associated with renal dysfunction: adverse events evaluation of treatment-naive patients undergoing front-line lenalidomide and dexamethasone therapy. Br. J. Haematol.138(5), 640–643 (2007).
  • Moreira AL, Sampaio EP, Zmuidzinas A, Frindt P, Smith KA, Kaplan G. Thalidomide exerts its inhibitory action on tumor necrosis factor alpha by enhancing mRNA degradation. J. Exp. Med.177(6), 1675–1680 (1993).
  • D’Amato RJ, Loughnan MS, Flynn E, Folkman J. Thalidomide is an inhibitor of angiogenesis. Proc. Natl Acad. Sci. USA91(9), 4082–4085 (1994).
  • Hideshima T, Chauhan D, Podar K, Schlossman RL, Richardson P, Anderson KC. Novel therapies targeting the myeloma cell and its bone marrow microenvironment. Semin. Oncol.28(6), 607–612 (2001).
  • Kumar S, Rajkumar SV. Thalidomide and lenalidomide in the treatment of multiple myeloma. Eur. J. Cancer42(11), 1612–1622 (2006).
  • Mitsiades N, Mitsiades CS, Poulaki V et al. Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: therapeutic implications. Blood99(12), 4525–4530 (2002).
  • Lepper ER, Smith NF, Cox MC, Scripture CD, Figg WD. Thalidomide metabolism and hydrolysis: mechanisms and implications. Curr. Drug Metab.7(6), 677–685 (2006).
  • Singhal S, Mehta J, Desikan R et al. Antitumor activity of thalidomide in refractory multiple myeloma. N. Engl. J. Med.341(21), 1565–1571 (1999).
  • Kneller A, Raanani P, Hardan I et al. Therapy with thalidomide in refractory multiple myeloma patients – the revival of an old drug. Br. J. Haematol.108(2), 391–393 (2000).
  • Juliusson G, Celsing F, Turesson I, Lenhoff S, Adriansson M, Malm C. Frequent good partial remissions from thalidomide including best response ever in patients with advanced refractory and relapsed myeloma. Br. J. Haematol.109(1), 89–96 (2000).
  • Tosi P, Zamagni E, Cellini C et al. Salvage therapy with thalidomide in patients with advanced relapsed/refractory multiple myeloma. Haematologica87(4), 408–414 (2002).
  • Arai A, Hirota A, Fukuda T et al. [Analysis of plasma concentration of thalidomide in Japanese patients of multiple myeloma with renal dysfunction]. Rinsho Ketsueki50(4), 295–299 (2009).
  • Seldin DC, Choufani EB, Dember LM et al. Tolerability and efficacy of thalidomide for the treatment of patients with light chain-associated (AL) amyloidosis. Clin. Lymphoma3(4), 241–246 (2003).
  • Tosi P, Zamagni E, Tacchetti P et al. Thalidomide-dexamethasone as induction therapy prior to autologous stem-cell transplantation in patients with newly diagnosed multiple myeloma and renal insufficiency. Biol. Blood Marrow Transplant16(8), 1115–1121 (2010).
  • Durie BG, Harousseau JL, Miguel JS et al. International uniform response criteria for multiple myeloma. Leukemia20(9), 1467–1473 (2006).
  • Blade J, Fernandez-Llama P, Bosch F et al. Renal failure in multiple myeloma: presenting features and predictors of outcome in 94 patients from a single institution. Arch. Intern. Med.158(17), 1889–1893 (1998).
  • Cavo M, Zamagni E, Tosi P et al. First-line therapy with thalidomide and dexamethasone in preparation for autologous stem cell transplantation for multiple myeloma. Haematologica89(7), 826–831 (2004).
  • Johnston RE, Abdalla SH. Thalidomide in low doses is effective for the treatment of resistant or relapsed multiple myeloma and for plasma cell leukaemia. Leuk. Lymphoma43(2), 351–354 (2002).
  • Fabro S, Smith RL, Williams RT. Toxicity and teratogenicity of optical isomers of thalidomide. Nature215(5098), 296 (1967).
  • Kastritis E, Anagnostopoulos A, Roussou M et al. Reversibility of renal failure in newly diagnosed multiple myeloma patients treated with high dose dexamethasone-containing regimens and the impact of novel agents. Haematologica92(4), 546–549 (2007).
  • Dimopoulos MA, Roussou M, Gavriatopoulou M et al. Reversibility of renal impairment in patients with multiple myeloma treated with bortezomib-based regimens: identification of predictive factors. Clin. Lymphoma Myeloma9(4), 302–306 (2009).
  • Matsue K, Fujiwara H, Iwama K, Kimura S, Yamakura M, Takeuchi M. Reversal of dialysis-dependent renal failure in patients with advanced multiple myeloma: single institutional experiences over 8 years. Ann. Hematol.89(3), 291–297 (2010).
  • Roussou M, Kastritis E, Christoulas D et al. Reversibility of renal failure in newly diagnosed patients with multiple myeloma and the role of novel agents. Leuk. Res.34(10), 1395–1397 (2010).
  • Clark WF, Stewart AK, Rock GA et al. Plasma exchange when myeloma presents as acute renal failure: a randomized, controlled trial. Ann. Intern. Med.143(11), 777–784 (2005).
  • Hutchison CA, Cockwell P, Reid S et al. Efficient removal of immunoglobulin free light chains by hemodialysis for multiple myeloma: in vitro and in vivo studies. J. Am. Soc. Nephrol.18(3), 886–895 (2007).
  • Hutchison CA, Bradwell AR, Cook M et al. Treatment of acute renal failure secondary to multiple myeloma with chemotherapy and extended high cut-off hemodialysis. Clin. J. Am. Soc. Nephrol.4(4), 745–754 (2009).
  • Hutchison CA, Cockwell P, Stringer S et al. Early reduction of serum-free light chains associates with renal recovery in myeloma kidney. J. Am. Soc. Nephrol.22(6), 1129–1136 (2011).
  • Adam Z, Krejci M, Tichy M, Stepankova S, Pour L, Hajek R. [The treatment of renal failure in multiple myeloma]. Vnitr. Lek.55(6), 570–582 (2009).
  • Sanders PW. A new twist in myeloma treatment. Blood107(2), 413–414 (2006).
  • Lacy MQ, Hayman SR, Gertz MA et al. Pomalidomide (CC4047) plus low-dose dexamethasone as therapy for relapsed multiple myeloma. J. Clin. Oncol.27(30), 5008–5014 (2009).
  • Richardson PG, Siegel D, Baz R et al. A Phase 1/2 multi-center, randomized, open label dose escalation study to determine the maximum tolerated dose, safety, and efficacy of pomalidomide alone or in combination with low-dose dexamethasone in patients with relapsed and refractory multiple myeloma who have received prior treatment that includes lenalidomide and bortezomib. ASH Annual Meeting Abstracts116(21), 864 (2010).
  • Richardson PG, Mitsiades C, Schlossman R, Munshi N, Anderson K. New drugs for myeloma. Oncologist12(6), 664–689 (2007).
  • Laubach JP, Mitsiades CS, Mahindra A et al. Novel therapies in the treatment of multiple myeloma. J. Natl Compr. Canc. Netw.7(9), 947–960 (2009).
  • Pasquali S, Zucchelli P, Casanova S et al. Renal histological lesions and clinical syndromes in multiple myeloma. Renal Immunopathology Group. Clin. Nephrol.27(5), 222–228 (1987).
  • Ivanyi B. Renal complications in multiple myeloma. Acta Morphol. Hung.37(3–4), 235–243 (1989).
  • Montseny JJ, Kleinknecht D, Meyrier A et al. Long-term outcome according to renal histological lesions in 118 patients with monoclonal gammopathies. Nephrol. Dial. Transplant.13(6), 1438–1445 (1998).
  • Herrera GA. Renal lesions associated with plasma cell dyscrasias: practical approach to diagnosis, new concepts, and challenges. Arch. Pathol Lab. Med.133(2), 249–267 (2009).
  • Uchiyama H, Barut BA, Mohrbacher AF, Chauhan D, Anderson KC. Adhesion of human myeloma-derived cell lines to bone marrow stromal cells stimulates interleukin-6 secretion. Blood82(12), 3712–3720 (1993).
  • Bartlett JB, Dredge K, Dalgleish AG. The evolution of thalidomide and its IMiD derivatives as anticancer agents. Nat. Rev. Cancer4(4), 314–322 (2004).
  • Yasui H, Hideshima T, Richardson PG, Anderson KC. Novel therapeutic strategies targeting growth factor signalling cascades in multiple myeloma. Br. J. Haematol.132(4), 385–397 (2006).
  • Multiple Myeloma: Translational and Emerging Therapies. Anderson KC, Ghobrial IM (Eds). Informa Healthcare, NY, USA (2007).

Management of myeloma-associated renal dysfunction in the era of novel therapies

To obtain credit, you should first read the journal article. After reading the article, you should be able to answer the following, related, multiple-choice questions. To complete the questions (with a minimum 70% passing score) and earn continuing medical education (CME) credit, please go to www.medscape.org/journal/experthematology. Credit cannot be obtained for tests completed on paper, although you may use the worksheet below to keep a record of your answers. You must be a registered user on Medscape.org. If you are not registered on Medscape.org, please click on the New Users: Free Registration link on the left hand side of the website to register. Only one answer is correct for each question. Once you successfully answer all post-test questions you will be able to view and/or print your certificate. For questions regarding the content of this activity, contact the accredited provider, [email protected]. For technical assistance, contact [email protected]. American Medical Association’s Physician’s Recognition Award (AMA PRA) credits are accepted in the US as evidence of participation in CME activities. For further information on this award, please refer to http://www.ama-assn.org/ama/pub/category/2922.html. The AMA has determined that physicians not licensed in the US who participate in this CME activity are eligible for AMA PRA Category 1 Credits™. Through agreements that the AMA has made with agencies in some countries, AMA PRA credit may be acceptable as evidence of participation in CME activities. If you are not licensed in the US, please complete the questions online, print the AMA PRA CME credit certificate and present it to your national medical association for review.

Activity Evaluation: Where 1 is strongly disagree and 5 is strongly agree

1. You are seeing a 70-year-old man with a 3-month history of fatigue and pain in his hips and low back. You note that he has had a 4-kg weight loss in the past 3 months. His serum creatinine level was above normal 2 months ago.

You consider whether this patient has multiple myeloma (MM) with renal insufficiency (RI). Which of the following statements regarding the diagnosis of MM and RI is most accurate?

  • A Patients with MM and RI featuring monoclonal immunoglobulin (Ig) deposition disease usually have hypotension

  • B Patients with MM and RI featuring amyloid deposition disease usually have hypertension

  • C Detection of serum or urine monoclonal Ig is no longer important in the diagnosis of MM

  • D Among patients with RI, elevated serum free light chain (SFLC) assay results may be misleading

2. The patient is diagnosed with both MM and RI and a renal biopsy is ordered. Which of the following histologic patterns is most common among patients with MM and RI?

  • A Amyloidosis

  • B Myeloma cast nephropathy

  • C Tubulointerstitial nephritis

  • D Acute tubular necrosis

3. Which of the following treatments would be considered first line for this patient?

  • A Lenalidomide

  • B Bortezomib

  • C Thalidomide

  • D Melphalan

4. What else should you consider regarding other therapeutic options for MM in the setting of RI?

  • A Lenalidomide does not require dose adjustment in the setting of RI

  • B High cut-off hemodialysis has been associated with renal recovery among patients receiving dialysis

  • C Thalidomide is associated with a lower rate of adverse events compared with bortezomib

  • D Melphalan is promising as a treatment in the setting of RI, but only at high doses

Notes

AH: Amyloid heavy chain amyloidosis; AL: Amyloid light chain amyloidosis.

Data taken from Citation[2,123–126].

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.