Patients with multiple myeloma (MM) frequently present with concomitant renal dysfunction, and those requiring dialysis have particularly poor outcomes. Bortezomib is a reversible proteasome inhibitor with significant activity in MM. This retrospective case analysis evaluated the feasibility and activity of bortezomib-based therapy in MM patients (n = 24) requiring dialysis support for advanced renal failure. All but 1 patient were undergoing dialysis at the time of therapy. Patients received bortezomib alone or bortezomib-based combination therapy. Among 20 patients with available response data, overall response rate (complete response [CR] + partial response) was 75%, with 30% CR + near CR. One patient was spared dialysis, and 3 other patients became independent of dialysis following bortezomib-based treatment. These encouraging results suggest that bortezomib or bortezomib-based regimens can be used in MM patients requiring dialysis, with manageable toxicities. Further studies will more formally evaluate the impact of bortezomib-based regimens in this patient population.

According to recent reports, approximately 30% of patients with diagnosed multiple myeloma (MM) present with baseline renal dysfunction,1–4  with 1% to 13% having renal failure requiring dialysis support.2–6  A number of studies have shown that the severity of renal impairment significantly affects the prognosis of patients with MM.1–4,6  Renal dysfunction has been associated with shorter survival or early death,1,4  posing challenges in delivering effective and safe treatment options.

Bortezomib is a first-in-class proteasome inhibitor indicated for the treatment of MM patients after 1 prior therapy. A recent subanalysis of patients with impaired renal function from two phase 2 studies showed that renal dysfunction did not appear to have a negative impact on response rates, toxicity, or treatment discontinuation in patients with relapsed and/or refractory MM receiving bortezomib therapy.7  To further examine the safety and activity of bortezomib-based therapy in MM patients with more severe renal impairment, we designed this retrospective analysis to include patients who required dialysis support at the time of bortezomib administration.

Consecutive patient cases were identified from 4 institutions in the United States experienced in myeloma treatment. The patient eligibility criteria included a diagnosis of MM, age at least 18 years, treatment with bortezomib alone or in combination with other agents, and renal failure requiring dialysis at the time of bortezomib treatment. Data on demographics, treatment schedule, response (by European Group for Blood and Marrow Transplantation [EBMT] criteria8 ), and adverse events were collected and analyzed using descriptive statistics. Patient consent was not required, because the collected data were existing information. The study protocol and data collection forms were approved by the institutional review board committees at all participating centers.

We have analyzed data collected from the records of 24 patients treated between May 30, 2003, and November 10, 2005. Patient demographics and baseline disease characteristics are provided in Table 1. All patients were scheduled to commence dialysis at the time of bortezomib administration. However, only 23 (96%) of 24 patients received dialysis, because 1 patient who rapidly responded to bortezomib therapy no longer required dialysis support (Document S1, available on the Blood website; see the Supplemental Document link at the top of the online article). Eighty-three percent of patients received bortezomib at a dose of 1.3 mg/m2 given in combination with other agents such as dexamethasone, thalidomide, and doxorubicin (Table 1). Patients received a median of 5 cycles (mean, 7 cycles) of bortezomib treatment.

Table 1

Demographics, pretreatment characteristics, and treatment schedule in 24 patients

CharacteristicsData
Male, % 50 
Median age, y (range) 59 (41-82) 
IgG/IgA/IgD/light chain only, % 29/25/4/42 
No. of prior therapies  
    Median (range) 2 (0-6) 
    None, no. (%) 1 (4) 
    1, no. (%) 4 (17) 
    2 or 3, no. (%) 12 (50) 
    4 or more, no. (%) 3 (13) 
    Unknown, no. (%) 4 (17) 
Serum creatinine level  
    Median, mg/dL (range) 6.8 (3.1-12.8) 
    More than 2 mg/dL, no. (%) 22 (92) 
    More than 6 mg/dL, no. (%) 15 (63) 
    Unknown, no. (%) 2 (8) 
Median hemoglobin level,* g/dL (range) 10.2 (7.4-15.3) 
Median platelet count,* × 109/L (range) 186 (25-369) 
Etiology of renal failure, no. (%)  
    Progressive MM 14 (58) 
    Amyloidosis 2 (8) 
    Renal cortical atrophy 1 (4) 
    Unknown 7 (29) 
Bortezomib treatment, no. (%)†  
    Alone 2 (8) 
    Plus dexamethasone 6 (25) 
    Plus dexamethasone and thalidomide 4 (17) 
    Plus liposomal doxorubicin and thalidomide 7 (29) 
    Other combinations 4 (17) 
    Not specified 1 (4) 
Bortezomib starting dose, no. (%)  
    Less than 1.0 mg/m2 1 (4) 
    1.0 mg/m2 3 (13) 
    1.3 mg/m2 20 (83) 
Median no. of cycles of bortezomib (range) 5 (2-20) 
Timing of treatment relative to dialysis, no. (%)  
    Before dialysis 1 (4) 
    During dialysis 1 (4) 
    After dialysis§ 20 (83) 
    Not reported 1 (4) 
    No dialysis 1 (4) 
CharacteristicsData
Male, % 50 
Median age, y (range) 59 (41-82) 
IgG/IgA/IgD/light chain only, % 29/25/4/42 
No. of prior therapies  
    Median (range) 2 (0-6) 
    None, no. (%) 1 (4) 
    1, no. (%) 4 (17) 
    2 or 3, no. (%) 12 (50) 
    4 or more, no. (%) 3 (13) 
    Unknown, no. (%) 4 (17) 
Serum creatinine level  
    Median, mg/dL (range) 6.8 (3.1-12.8) 
    More than 2 mg/dL, no. (%) 22 (92) 
    More than 6 mg/dL, no. (%) 15 (63) 
    Unknown, no. (%) 2 (8) 
Median hemoglobin level,* g/dL (range) 10.2 (7.4-15.3) 
Median platelet count,* × 109/L (range) 186 (25-369) 
Etiology of renal failure, no. (%)  
    Progressive MM 14 (58) 
    Amyloidosis 2 (8) 
    Renal cortical atrophy 1 (4) 
    Unknown 7 (29) 
Bortezomib treatment, no. (%)†  
    Alone 2 (8) 
    Plus dexamethasone 6 (25) 
    Plus dexamethasone and thalidomide 4 (17) 
    Plus liposomal doxorubicin and thalidomide 7 (29) 
    Other combinations 4 (17) 
    Not specified 1 (4) 
Bortezomib starting dose, no. (%)  
    Less than 1.0 mg/m2 1 (4) 
    1.0 mg/m2 3 (13) 
    1.3 mg/m2 20 (83) 
Median no. of cycles of bortezomib (range) 5 (2-20) 
Timing of treatment relative to dialysis, no. (%)  
    Before dialysis 1 (4) 
    During dialysis 1 (4) 
    After dialysis§ 20 (83) 
    Not reported 1 (4) 
    No dialysis 1 (4) 
*

Data available from 22 patients.

Bortezomib was administered using the standard dose schedule (days 1, 4, 8, and 11 every 3 weeks) except in 7 patients who received an alternative schedule of bortezomib (days 1, 4, 15, and 18 every 4 weeks).

Other regimens included combinations with cyclophosphamide, doxorubicin, thalidomide, dexamethasone, and prednisone.

§

One patient received treatment both after and before the dialysis procedure on separate hospital visits.

Most adverse events were graded mild to moderate and were manageable (Table 2; n = 18). As expected for bortezomib-based therapy, the most common adverse event was reversible thrombocytopenia (39%), with no bleeding events. Mean platelet levels remained at least 150 × 109/L at the time of initiation of each cycle (ie, day 1) of bortezomib treatment (n = 15). Among the 18 patients for whom data were available, 8 patients (44%) discontinued therapy (Table 2). One patient died because of progressive disease.

Table 2

Summary of adverse events (all grades) and treatment discontinuation in 18 patients

No. (%)
Adverse events  
    Neuropathic pain 1 (6) 
    Peripheral neuropathy 2 (11) 
    Neutropenia 1 (6) 
    Headache 1 (6) 
    Hypertension 1 (6) 
    Infections 2 (11) 
    Thrombocytopenia 7 (39) 
    Platelet count 50 × 109/L to less than 150 × 109/L 5 (28) 
    Platelet count 25 × 109/L to less than 50 × 109/L 2 (11) 
Serious adverse events  
    Death due to progressive disease 1 (6) 
Adverse events leading to discontinuation  
    Progressive disease 6 (33) 
    Neuropathic pain 1 (6) 
    Peripheral neuropathy 1 (6) 
No. (%)
Adverse events  
    Neuropathic pain 1 (6) 
    Peripheral neuropathy 2 (11) 
    Neutropenia 1 (6) 
    Headache 1 (6) 
    Hypertension 1 (6) 
    Infections 2 (11) 
    Thrombocytopenia 7 (39) 
    Platelet count 50 × 109/L to less than 150 × 109/L 5 (28) 
    Platelet count 25 × 109/L to less than 50 × 109/L 2 (11) 
Serious adverse events  
    Death due to progressive disease 1 (6) 
Adverse events leading to discontinuation  
    Progressive disease 6 (33) 
    Neuropathic pain 1 (6) 
    Peripheral neuropathy 1 (6) 

The overall response rate was 75% among the 20 patients with response data. Five patients had a complete response (CR; 25%), 1 a near CR (meeting EBMT criteria for CR but having a positive immunofixation test result; 5%), and 9 a partial response (PR; 45%). In addition, 1 patient (5%) had a minimal response and 2 other patients (10%) had stable disease. Among 10 responding patients for whom data were available, the median duration of response was 12.5+ months (range, 2.5 to 20+ months). The median duration of CR was 15 months (range, 6 to 20+ months), and the median duration of PR was 4.5 months (range, 2.5 to 13 months). The duration of response for the 1 patient achieving near CR was 12+ months. A total of 4 patients had improved renal function following bortezomib-based therapy (Document S1), including 1 patient who was spared dialysis after a rapid response to treatment, 2 patients who no longer required dialysis support after achieving a CR, and a fourth patient who became independent of dialysis following a minimal response to therapy. In the case of the fourth patient, normalization of renal function may not have been primarily attributable to bortezomib-based therapy.

At the present time, no standard guidelines have been established to treat MM patients with renal failure requiring dialysis. The presence of concomitant renal dysfunction has been considered a negative prognostic indicator associated with decreased response,2  shorter survival,2,4  and early mortality1,6  in patients treated with conventional chemotherapy. Patients with irreversible renal dysfunction or requiring dialysis have a particularly poor prognosis, with a median survival of less than 4 months.2,3  Treatment options such as melphalan-based chemotherapy3,6,9  and high-dose chemotherapy with autologous stem cell transplantation5,10–12  have limitations in this patient population because of suboptimal survival benefits, excessive toxicities, early mortality, and/or the need for dose reductions and treatment discontinuations. The recent introduction of targeted agents such as thalidomide (or the thalidomide analog, lenalidomide) has improved outcomes for MM patients. However, there is limited information regarding their safety and activity in the subgroup of high-risk MM patients with dialysis-dependent renal failure. Thalidomide has been associated with a high incidence of severe and potentially fatal hyperkalemia, particularly in patients undergoing hemodialysis.13,14  Lenalidomide has been shown to undergo substantial elimination via the kidneys and may therefore be associated with greater toxicities in patients with impaired renal function. In fact, patients with significant renal insufficiency were routinely excluded from clinical trials; postmarketing studies of lenalidomide in MM patients with renal dysfunction are currently underway.

Overall, toxicities were manageable and similar to those seen in patients with relapsed and/or refractory MM enrolled in the pivotal phase 2 SUMMIT15  and CREST16  trials, the phase 3 APEX17  trial, and other studies evaluating bortezomib-based therapies in previously treated MM.18–20  Given the retrospective nature of the analysis, the safety data for our study may have been underreported; nevertheless, the results from available data appear encouraging. Moreover, the patients in this study were able to receive a median of 5 cycles (or mean of 7 cycles) of bortezomib-based therapy, comparable to previous reports in patients with normal renal function.15,16 

The overall response rate and durability of response to bortezomib-based treatments in our dialysis-dependent patients appear comparable to those of MM patients with primarily normal renal function reported in other studies in the relapsed setting.15–21  In our study, 83% of patients received treatment after completion of dialysis, suggesting that delivery of bortezomib subsequent to dialysis does not appear to affect the activity of the drug. The present study also suggests that bortezomib treatment has a potentially positive impact on renal function, with normalization of renal dysfunction in a few patients. These observations are consistent with recent reports from other investigators who noted improvement in renal function22  as well as complete or near-complete reversal of severe renal dysfunction21,23,24  following bortezomib-based combination therapy.

Our study suggests that bortezomib therapy is a well-tolerated, effective option in the subgroup of MM patients with severe renal dysfunction. Preliminary data from the prospective National Cancer Institute pharmacologic trial showed that bortezomib clearance is independent of renal function, and the standard 1.3 mg/m2 dose appeared well tolerated in patients with mild to moderate renal dysfunction.25  Accrual continues for patients having more severe renal dysfunction or requiring dialysis, and further analysis of the results from this study will help characterize the safety profile of bortezomib in this group of patients.

Contribution: A.A.C.-K. wrote the paper, participated in the scientific design, and conducted research and patient accrual; J.L.K, J.M., P.G.R., K.C.M., S.L., N.C.M., R.S., J.T., and S.S. conducted patient accrual; and all authors checked the final version of the manuscript.

Conflict-of-interest disclosure: P.G.R., K.C.M., S.L., N.C.M., R.S., and J.T. have no financial relationships to disclose. A.C.K., J.M., and S.S. are members of the speakers bureau for Millennium Pharmaceuticals, Inc. J.L.K. is a consultant for Millennium Pharmaceuticals, Inc. A.C.K. is a member of the advisory board for Millennium Pharmaceuticals, Inc. J.M. is a member of the speakers bureau for Celgene Corporation.

Correspondence: Asher Chanan-Khan, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY 14263; e-mail: asher.chanan-khan@roswellpark.org.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

1
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
2005
;
23
:
9219
–9226.
2
Bladé 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
1998
;
158
:
1889
–1893.
3
Knudsen LM, Hjorth M, Hippe E, et al. Renal failure in multiple myeloma: reversibility and impact on the prognosis.
Eur J Haematol
2000
;
65
:
175
–181.
4
Kyle RA, Gertz MA, Witzig TE, et al. Review of 1027 patients with newly diagnosed multiple myeloma.
Mayo Clin Proc
2003
;
78
:
21
–33.
5
Knudsen LM, Nielsen B, Gimsing P, et al. Autologous stem cell transplantation in multiple myeloma: outcome in patients with renal failure.
Eur J Haematol
2005
;
75
:
27
–33.
6
Torra R, Blade J, Cases A, et al. Patients with multiple myeloma requiring long-term dialysis: presenting features, response to therapy, and outcome in a series of 20 cases.
Br J Haematol
1995
;
91
:
854
–859.
7
Jagannath S, Barlogie B, Berenson JR, et al. Bortezomib in recurrent and/or refractory multiple myeloma, Initial clinical experience in patients with impaired renal function.
Cancer
2005
;
103
:
1195
–1200.
8
Blade J, Samson D, Reece D, et al. Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplanatation.
Br J Haematol
1998
;
102
:
1115
–1123.
9
Vigneau C, Ardiet C, Bret M, et al. Intermediate-dose (25mg/m2) IV melphalan for multiple myeloma with renal failure.
J Nephrol
2002
;
15
:
684
–689.
10
Badros A, Barlogie B, Siegel E, et al. Results of autologous stem cell transplant in multiple myeloma patients with renal failure.
Br J Haematol
2001
;
114
:
822
–829.
11
Carlson K. Melphalan 200 mg/m2 with blood stem cell support as first-line myeloma therapy: impact of glomerular filtration rate on engraftment, transplantation-related toxicity and survival.
Bone Marrow Transplant
2005
;
35
:
985
–990.
12
Lee CK, Zangari M, Barlogie B, et al. Dialysis-dependent renal failure in patients with myeloma can be reversed by high-dose myeloablative therapy and autotransplant.
Bone Marrow Transplant
2004
;
33
:
823
–828.
13
Fakhouri F, Guerraoui H, Presne C, et al. Thalidomide in patients with multiple myeloma and renal failure.
Br J Haematol
2004
;
125
:
96
–97.
14
Harris E, Behrens J, Samson D, et al. Use of thalidomide in patients with myeloma and renal failure may be associated with unexplained hyperkalaemia.
Br J Haematol
2003
;
122
:
160
–161.
15
Richardson PG, Barlogie B, Berenson J, et al. A phase 2 study of bortezomib in relapsed, refractory myeloma.
N Engl J Med
2003
;
348
:
2609
–2617.
16
Jagannath S, Barlogie B, Berenson J, et al. A phase 2 study of two doses of bortezomib in relapsed or refractory myeloma.
Br J Haematol
2004
;
127
:
165
–172.
17
Richardson PG, Sonneveld P, Schuster MW, et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma.
N Engl J Med
2005
;
352
:
2487
–2498.
18
Orlowski RZ, Voorhees PM, Garcia RA, et al. Phase 1 trial of the proteasome inhibitor bortezomib and pegylated liposomal doxorubicin in patients with advanced hematologic malignancies.
Blood
2005
;
105
:
3058
–3065.
19
Kropff M, Bisping G, Liebisch P, et al. Bortezomib in combination with high-dose dexamethasone and continuous low-dose oral cyclophosphamide for relapsed multiple myeloma [abstract].
Blood
2005
;
106
:
716a
Abstract 2549.
20
Zangari M, Barlogie B, Elice F, et al. Marked activity of VTD regimen comprising Velcade (V) plus thalidomide (T) and added dexamethasone for non-responders in advanced and refractory multiple myeloma (MM) [abstract].
Haematologica
2005
;
90
:
255
Abstract 637.
21
Chanan-Khan A and Miller KC. Velcade, Doxil and Thalidomide (VDT) is an effective salvage regimen for patients with relapsed and refractory multiple myeloma.
Leuk Lymphoma
2005
;
46
:
1103
–1104.
22
Nozza A, Siracusano L, Armando S. Bortezomib-dexamethasone combination in a patient with refractory multiple myeloma and impaired renal function.
Clin Ther
2006
;
28
:
953
–959.
23
Ludwig HL, Zojer N, Kuenburg E, et al. Reversal of acute multiple myeloma (MM)-induced renal failure by bortezomib-combination therapy: a report of three cases [abstract].
Haematologica
2005
;
90
:suppl 2,
430
Abstract 1112.
24
Mohrbacher A and Levine AM. Reversal of advanced renal dysfunction on bortezomib treatment in multiple myeloma patients [abstract].
J Clin Oncol
2005
;
23
:suppl 16,
612s
Abstract 6714.
25
Mulkerin D, Remick S, Ramanathan R, et al. A dose-escalating and pharmacologic study of bortezomib in adult cancer patients with impaired renal function [abstract].
J Clin Oncol
2006
;
24
:
18S
Abstract 2032.
Sign in via your Institution