Abstract
Bortezomib is the first therapeutic inhibitor of the proteasome that has demonstrated a significant clinical response in patients with otherwise refractory or rapidly advancing disease. Bortezomib has received US Federal Drug Administration approval for the treatment of the hematologic malignancies such as multiple myeloma and mantle cell lymphoma. Herein, the use of bortezomib as an upfront therapy, as an induction regimen before stem-cell transplantation and as maintenance therapy in the treatment of multiple myeloma is discussed.
Keywords: multiple myeloma, bortezomib, monotherapy, stem-cell transplantation induction, maintenance therapy
INTRODUCTION
Multiple myeloma (MM) is a plasma cell proliferative disorder characterized by the uncontrolled growth and dysfunction of the malignant cells within the bone marrow.1,2 MM is the second most common hematological cancer worldwide and accounts for >11,000 deaths each year in the United States.3,4 Although MM is predominantly a disease of the elderly with an average age of onset of 65–70 years, recent statistics indicate both increasing incidence and younger age of onset. In the United States, >50,000 individuals have MM, and 20,000 new cases are diagnosed each year, whereas worldwide, there are ~74,000 new cases and >45,000 deaths annually. MM remains an incurable disease, and novel treatment approaches are therefore urgently needed to alleviate patient symptoms and to improve overall survival (OS).
Therapy for MM simultaneously attempts to reduce or eradicate existing disease while also aiming to control disease-related hematologic, bone, and renal complications.5 The main treatment modalities include cytotoxic chemotherapy to control disease burden and rarely leads to complete remission; corticosteroids such as dexamethasone and prednisone, immunomodulatory agents such as thalidomide and lenalidomide used in newly diagnosed, those with advanced disease who have failed chemotherapy or stem-cell transplantation (SCT) and new biologic agents such as the proteasome inhibitor bortezomib. In most cases, these agents are used in combination with standard chemotherapy agents. For many years, melphalan and prednisone remained the standard therapy for MM, and response rates (RRs) were ~50% with median survival of ~3 years.6 Recently, autologous SCT has demonstrated promising results in randomized clinical trials as well.7,8 Patients eligible for SCT typically avoid alkylator-based induction therapy to enable collection of an adequate and functionally viable harvest of stem cells.9 As initial therapy in newly diagnosed or relapsed patients, SCT is generally not curative but has been shown to prolong survival.7,8 Additionally, in selected patients, multiple SCTs may be employed. Advanced age and risk factors such as increased β2-microglobulin levels, low serum albumin levels, abnormal cytogenetics, including chromosome 13 deletion, and being refractory to prior treatment are associated with poorer prognosis in MM and may limit treatment options.10–12 Depending on the treatment regimen used, patients with renal impairment may achieve lower RRs and shorter OS, and there may be greater safety concerns.
Bortezomib is a synthetic, boronyl dipeptide that highly selectively and reversibly inhibits the proteasome and has multiple effects on MM cell lines and primary human MM cells.13–15 Bortezomib may inhibit the tightly regulated turnover of a number of proteins that malignant plasma cells need for cellular growth and proliferation. Normal, noncancerous cells are not as susceptible to the deleterious effects of bortezomib as are malignant cells. Bortezomib was approved in May 2003 by the US Federal Drug Administration (FDA) for the treatment of patients with relapsed or refractory MM and who had received at least 2 prior lines of therapy and progressed on their last therapy.16,17 In 2005, bortezomib was approved by the FDA for the treatment of patients with MM who had received at least 1 prior therapy.18 In December 2006, the FDA approved bortezomib for treatment of mantle cell lymphoma (MCL).19 Finally, on June 20, 2008, the FDA approved bortezomib for injection as the upfront treatment of patients newly diagnosed with MM based upon an international, multicenter, open-label, active-control trial in previously untreated patients with symptomatic MM.20 Time-to-progression (TTP) was the primary efficacy end point, whereas OS, progression-free survival (PFS), and RR were the secondary endpoints. Patients were randomized to receive either melphelan + prednisone (MP) or bortezomib + melphelan + prednisone. The trial was stopped after a prespecified interim analysis showing a statistically significant improvement in TTP with the addition of bortezomib to MP (median: 20.7 months) compared with MP (median: 15 months), whereas OS, PFS, and RR also were significantly superior for the bortezomib–MP combination. Bortezomib is generally well tolerated with mild-to-moderate side effects that are manageable and include peripheral neuropathy and thrombocytopenia. The benefit of a bortezomib-based therapy in MM and MCL, diffuse large B-cell lymphoma and the plasma cell dyscrasias Waldenstroms Macro-globulinemia and Systemic Amyloidosis has been recently reviewed.21
BORTEZOMIB AS UPFRONT THERAPY IN MULTIPLE MYELOMA
The objective of frontline therapy in most hematologic malignancies is to maximally kill tumor cells to reduce tumor burden in preparation for consolidation therapy with SCT or as a means in itself to generate long-term disease control. In MM, such a therapy is usually followed by high-dose melphalan and autologous SCT. Based upon promising preclinical studies, the combination of bortezomib with oral dexamethasone demonstrated substantial response in the treatment of relapsed MM shown to be refractory to conventional chemotherapy.17 The RR to bortezomib was 35%, and median OS was 16 months with a duration of 12 months. Subsequently, bortezomib was compared to high-dose dexamethasone in MM patients with relapsed disease and who had received 1–3 prior therapies.18 Bortezomib was superior to high-dose dexamethasone with complete response (CR) and partial response (PR) total of 38% vs 18% for DEX and median TTP of 6.2 vs 3.5 months. To assess efficacy and safety of single-agent bortezomib in previously untreated patients with multiple myeloma (MM), San Miguel et al20 investigated the prevalence of baseline and treatment-emergent polyneuropathy and identified molecular markers associated with response and neuropathy. Among 64 patients, 41% had partial response or better, including 9% complete/near-complete responses; the median duration of response was 8.4 months. RRs did not differ in the presence or absence of adverse cytogenetics. After a median follow-up of 29 months, median time to progression was 17.3 months. Single-agent bortezomib is effective in previously untreated myeloma. Baseline myeloma-associated neuropathy seems more common than previously reported, and bortezomib-associated neuropathy, although a common toxicity, is reversible in most patients. An interesting approach was used by Dispenzieri et al22 using single-agent bortezomib as induction, maintenance, and reinduction in patients with high-risk MM. Eastern Cooperative Oncology Group (ECOG) trial E2A02 studied single-agent bortezomib and prospectively targeted high-risk patients and was not followed by consolidative autologous SCT. Moreover, in contrast to other studies, patients were not slated for a set number of cycles or therapy followed by high-dose chemotherapy ± peripheral SCT. Among high-risk patients, single-agent bortezomib induced a 51% ORR, which was comparable to that reported by others in unselected patient populations. A limitation in the study was that patients with t(4:14) were underrepresented. Although bortezomib was equally efficacious in high-risk and low-risk populations, it does not seem to be the optimal treatment choice to achieve sustained disease free survival in the absence of SCT or high-dose chemotherapy.
BORTEZOMIB AS INDUCTION THERAPY IN STEM-CELL TRANSPLANTATION FOR MULTIPLE MYELOMA
Autologous SCT is considered the gold standard initial therapy for patients <65 years of age who have been diagnosed with MM, because it results in higher rates of CR and longer time of event-free survival.23 The incorporation of novel agents has resulted in higher pre and posttransplantation CR rates, and induction with bortezomib-containing regimens has shown promising results in those with poor-risk cytogenetics. Conventional induction regimens followed by single or double autologous stem-cell transplantation (ASCT) result in a median survival of 6 years with immunofixation negative CR rates up to 40% under optimal circumstances.23–25 The combination of thalidomide + dexamethasone (THAL-DEX) is the current FDA approved pretransplantation induction regimen, but numerous strategies that incorporate novel agents such as bortezomib have been and are being investigated. In 2005, Jagannath et al26 reported results of a phase 2 trial that used the combination of bortezomib + dexamethasone for the treatment of 32 newly diagnosed patients with MM (Table 1). Patients received bortezomib 1.3 mg/m2 intravenous (IV) on days 1, 4, 8, and 11 every 3 weeks for a maximum of 6 cycles. Twenty-two patients went on to receive dexamethasone (40 mg) added to their regimen on the day of and day after bortezomib administration because they had less than a PR after 2 cycles or less than a CR after 4 cycles. There was an overall response rate (ORR) of 88%, and most patients then received high-dose chemotherapy with peripheral SCT. Harousseau et al27 used bortezomib + dexamethasone as induction in 48 patients and achieved an ORR of 66%. Treatment regimen consisted of bortezomib 1.3 mg/m2 IVon days 1, 4, 8, and 11 and 40 mg of dexamethasone on days 1–4 and 9–12 during cycles 1 and 2 and reduced to days 1–4 during cycles 3 and 4. Patients were treated for 4–21 day cycles. Rosinol et al28 reported newly diagnosed MM patients with 6 alternating cycles of bortezomib and dexamethasone and 60% ORR. The regimen used was bortezomib 1.3 mg/m2 IV on days 1, 4, 8, and 11 alone during cycles 1, 3, and 5 and with dexamethasone (40 mg) on days 1–4, 9–12, 17–20 during cycles 2, 4, and 6. Richardson et al29 reported treatment of 64 MM patients with single-agent bortezomib and an ORR of 41%, but 32 underwent peripheral stem cell transplant. Importantly, nearly half the patients in this study did not go on to receive SCT, and extended analysis may offer uncomplicated PFS and OS information.
Table 1.
Reference | FDA/NCCN | Patient population |
Patients | Regimen | Best response | Survival data |
---|---|---|---|---|---|---|
Jagannath et al26 | N/Y – (Cat 1) | Previously untreated | 32 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 6–3 wk cycles | 88% ORR | OS 1 yr—87% |
Dex (40 mg/d of/after) for ≤PR after 2 cycles; ≤CR after 4 cycles | 25% CR/nCR | |||||
Harousseau et al27 | N/Y – (Cat 1) | Previously untreated | 48 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 4–3 wk cycles | 66% ORR | NR |
Dex 40 mg on D1–4, 9–12 on C1 & 2; D1–4 on C3 & 4 | 21% CR | |||||
Consolidation to auto-SCT | 10% VGPR | |||||
Rosinõl et al28 | N/Y – (Cat 1) | Previously untreated | 40 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 6–3 wk cycles | 77.5% ORR | NR |
Dex 40 mg on D1–4, 9–12, 17–20 on C2, 4, & 6 | 12.5% CR | |||||
Consolidation to auto-SCT | 7.5% VGPR | |||||
Richardson et al29 | N/N | Previously untreated | 64 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 | 63% ORR | Med PFS—17 mos |
Max 8–3 wk cycles or 2 cycles past CR | 9% CR/nCR | OS 2.5 yrs—79% | ||||
8% VGPR | ||||||
Dispenzieri et al22 | N/N | Previously untreated | 39 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 8–3 wk cycles | 51% ORR | PFS 2 yrs—16% |
High risk per protocol | Maintenance D1, 15 of 28 d cycle | 0% CR | OS 2 yrs—76% | |||
Reinduction at PD | 10% VGPR | |||||
Popat et al30 | Y/Y – (Cat 1) | Previously untreated | 41 | PAD1 (phase I) | 62% ≥VGPR | PFS—29 v 24 mos |
–Induction regimen | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 4–3 wk cycles | 42% ≥VGPR | OS 1 yr−100% v 95% | |||
–Phase I/II | Doxo 0, 4.5, 9 mg/m2 on D1–4 to max admin dose | (PAD1 v 2 – induction) | ||||
Dex 40 mg on D1–4, 8–11, 15–18—C1, D1–4—C2–4 PAD2 (phase II) | ||||||
Bor 1 mg/m2 on D1, 4, 8, & 11 for 4–3 wk cycles | 81% ≥VGPR | OS 2 yrs | ||||
Doxo 9 mg/m2 on D1–4 | 53% ≥VGPR | −95% v 73% | ||||
Dex 40 mg D1–4, 8–11, 15–18—C1, D1–4—C2–4 | (PAD1 v 2 – posttransplant) | |||||
Consolidation w/auto-SCT | ||||||
Cavo et al31 | N/Y – (Cat 1) | Previously untreated | 474 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 3–3 wk cycles | 19% CR* | PFS 2 yrs—85%* |
Induction regimen | Thal 200 mg/d on D1–63 | 62% ≥ VGPR* | OS 20 mos—93% | |||
Phase III trial | Dex 40 mg D1, 2, 4, 5, 8, 9, 11, 12 | Postinduction | ||||
Consolidation w/auto-SCT × 2→ VTD × 2- 35 d cycle | 44% CR* | |||||
Compared to TD→ auto-SCT × 2→ TD × 2- 35 d cycle | 80% ≥ VGPR* | |||||
Full treatment | ||||||
Palumbo et al32 | N/N | Previously untreated | 102 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 4– 3 wk cycles | 13% CR | PFS 2 yrs—69% |
Induction regimen | PLD 30 mg/m2 on D4 | 58% ≥VGPR | OS 2 yrs—86% | |||
Phase II | Dex 40 mg D1–4, 8–11, 15–18—C1, D1–4—C2–4 | Postinduction | ||||
Consolidation w/tandem auto-SCT w/MEL100 | 38% CR | |||||
Followed by Len/Pred→ maintenance Len | 82% ≥VGPR | |||||
Posttransplant | ||||||
Moreau et al33 | N/N | Previously untreated | 205 | Bor 1 mg/m2 on D1, 4, 8, & 11 for 4–3 wk cycles | 31% CR/nCR | NR |
Induction regimen | Thal 100 mg/d on D1–21 | 50% ≥ VGPR* | ||||
Phase III trial | Dex 40 mg D1–4, 8–11 | Postinduction | ||||
Consolidation w/auto-SCT | 60% CR/nCR | |||||
Compared to standard VTD regimen above | 66% ≥ VGPR* | |||||
Post 1st ASCT | ||||||
Roussel et al34 | N/N | Nonprogressive MM after induction | 53 | Bor 1 mg/m2 on D-6, −3, +1, +4 | 32% CR | Median PFS-NR |
Conditioning reg. | MEL200 on D-2 | 70% ≥VGPR | PFS 2 yrs—76% | |||
Phase II trial | Auto-SCT reinfusion on D0 | Posttransplant | ||||
Harousseau et al35 | N/Y – (Cat 1) | Previously untreated | 482 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 4–3 wk cycles | 15% CR/nCR* | PFS—36 mos |
Induction regimen | Dex 40 mg D1–4, 9–12 on C1 & 2; D1–4 on C3 & 4 | 38% ≥ VGPR* | OS 3 yrs—81% | |||
Phase III trial | Consolidation w/auto-SCT ± DCEP | 79% ORR* | ||||
Compared to VAD induction | Postinduction | |||||
35% CR/nCR* | ||||||
54% ≥ VGPR* | ||||||
Post 1st ASCT | ||||||
Bruno et al36 | N/N | Relapse s/p AlloSCT | 23 | Bor 1–1.3 mg/m2 on D1, 4, 8, & 11 4-wk cycles | 61% ORR | PFS—6 mos |
Retrospective | ±Dex 20–40 mg on D1, 4, 15, and 18 | 22% CR | OS 6 mos—91% | |||
Kröger et al37 | N/N | Post-allo-SCT | 18 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 2–3 wk cycles | 30% CR | NR |
Enhance/maintain remission | 50% PR | |||||
20% Min Res (10 patients w/residual dz) | ||||||
Kröger et al38 | N/N | Post-allo-SCT | 8 | Bor 1.3 mg/m2 on D1, 4, 8, & 11 for 4–3 wk cycles | 59% CR | PFS 5 yrs—53% |
DLI + Bor/Thal/Len | Patients received median of 2 cycles (1–5) | OS 5 yrs—90% | ||||
(no specific Bor data mentioned) | ||||||
Sonneveld et al39 | N/N | Maintenance | 833 | Bor 1.3 mg/m2 every 2 wks for 2 yrs | CR 15→ 27% | NR |
Phase III trial | VAD v. PAD induction→ auto-SCT × 1–2→ Thal 50 mg/d (VAD arm) vs. Bor (PAD arm) | |||||
Rosinõl et al40 | N/N | Maintenance | 390 | Bor/Thal Maintenance for planned 3 yrs | NR | NR |
No doses noted in literature/clinical trials.gov | ||||||
Compared to Thal alone and IFN | ||||||
Palumbo et al41 | N/N | Maintenance | 511 | Bor/Mel/Pred/Thal induction | Maintenance did not improve RR | NR |
Bor 1.3 mg/m2 every 2 wks | ||||||
Thal 50 mg/d | ||||||
Ladetto et al42 | N/N | Consolidation/maintenance | 40 | Bor 1.6 mg/m2 on D1, 8, 15, 22 for 4–5 wk cycles | CR 15→ 49% | PFS—60 mos |
Thal 50 mg/d→ 200 mg/d; increase weekly as tolerated | MR 3→ 18% | OS 3 yrs—89% | ||||
Dex 20 mg D1–4, 8–11, 15–18 | ||||||
Nair et al25 | N/N | Maintenance | 177 | Bor 1 mg/m2 on D1, 4, 8, & 11 monthly for 1 yr | NR | NR |
Bor 1 mg/m2 weekly for 2 yrs | ||||||
Combined with Len/Dex for planned all 3 yrs | ||||||
Kumar et al43 | N/N | Maintenance | 117 | Bor 1.3 mg/m2 on D1, 8, 15, & 22 for 4–42 d cycles | NR | NR |
Richardson et al44 | N/N | Maintenance | 68 | Bor on D1 & 8 every 3 wks | NR | NR |
Combined with Len/Dex | ||||||
No duration noted in article | ||||||
Mateos et al45 | N/N | Maintenance | 260 | Bor/Thal/Pred v Bor/Mel/Pred Induction | CR | NR |
Bor 1.3 mg/m2 on D1, 4, 8, & 11—3-wk cycle | VP: 39% | |||||
Given every 3 mos for 3 yrs | VT: 44% | |||||
Plus either Pred/Thal |
Statistically significant difference between control and treatment groups
DLI, donor lymphocyte infusion.
The Intergroupe Francophone Du Myelome 2005-01 trial was conducted with 482 patients and demonstrated statistically significant differences in CR/near-CR (nCR) rates (21% vs 8%, P = 0.0023) and ≥VGPR rates (47% vs 19%) to favor the VD (bortezomib + dexamethasone) regimen.35 The benefit of the bortezomib + dexamethasone arm was maintained after the first auto-SCT. In addition, stem cells were mobilized from recipients who received bortezomib-based induction without additional complications. An update was presented, and the benefit of the bortezomib-based regimen was maintained.46 Survival results were presented, and though PFS was not significant between the 2 groups (36 vs 30 months), this changed if patients in both cohorts achieved ≥VGPR with induction (41 vs 29 months; P < 0.001) or achieved ≥VGPR with transplant (41 vs 33 months; P = 0.0257). There was no detectable significant difference in the VD group between patients with advanced stage or poor cytogenetics, although the analysis of patient cytogenetics was not exhaustive. A modified induction regimen of bortezomib/thalidomide/dexamethasone (VTD) using a reduced bortezomib dose of 1.0 mg/m2 and thalidomide of 100 mg/d (vTD) showed a significant CR + VGPR improvement for the experimental arm with improvement in peripheral neuropathy.47 An update on this study as well indicated that the median PFS had not been reached, and with a 2-year PFS of 76%, there was no difference from the 3 induction groups. However, patients who received VD induction, VEL–MEL conditioning for auto-SCT and consolidation had a 2-year PFS of 100%.31
Because bortezomib in combination with pegylated, liposomal doxorubicin (PLD) was shown to have significant activity against relapsed/refractory MM, this combination was evaluated in previously untreated MM patients who required induction chemotherapy before SCT. The combination of bortezomib and PLD was well tolerated by chemotherapy-naïve patients, and the steroid-free regimen, as seen previously with bortezomib-containing combinations, did not compromise the collection of adequate stem-cell populations for transplantation.48 The bortezomib-containing regimens TT2 and TT3 have been extensively well characterized and have shown success as well.49 TT3 used abbreviated induction and consolidation therapies with 2 rather than 4 cycles each in TT2, on the assumption that the addition of bortezomib (V) to DT-PACE (dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide, etoposide) in VDT-PACE would be highly synergistic so that comparable antitumor activity would be delivered with fewer cycles. Drug-free phases of TT2 were ‘bridged’ by THAL-DEX in an effort to suppress the potentially MM-stimulatory signals associated with postchemotherapy hematopoietic recovery. Bortezomib was then combined with THAL-DEX in VTD, which was applied in monthly cycles during the first year of maintenance, followed by THAL-DEX for 2 more years. In the case of the newer agent combinations such as MPT (melphalan, prednisone, thalidomide) or MPT with added bortezomib, greater compliance with the intended therapies is at least partially responsible for their unexpected high success rate. Bortezomib, doxorubicin and dexamethasone (PAD) was evaluated as induction before SCT in newly diagnosed MM patients, using bortezomib 1.3 mg/m2 (PAD1, N = 21) or 1.0 mg/m2 (PAD2, N = 20).30 CR/VGPR rates with PAD1/PAD2 were 62%/42% postinduction and 81%/53% posttransplant. PFS (29 vs 24 months), time to retreatment (36 vs 29 months) and OS (1 year: 100% vs 95%; 2 years: 95% vs73%) were statistically similar but favored PAD1 vs PAD2. Toxicity was lower in PAD2; bortezomib dose reduction may help manage toxicities while retaining efficacy. PAD is highly active as frontline induction in MM.
Preclinical studies have demonstrated that bortezomib has no toxic effects on stem cells, megakaryocytes, or neutrophil precursors and causes only transient and reversible thrombocytopenia and neutropenia.50 Numerous clinical studies with bortezomib-based induction regimens have demonstrated no adverse impact on peripheral blood stem cell harvest numbers nor on their quality as defined by engraftment times. These regimens seem to be well tolerated and highly active as induction therapy, with high RRs and consistently high CR rates. The standard treatment for patients with MM who are not candidates for high-dose therapy is melphalan and prednisone.51 The phase 3 study compared the use of melphalan and prednisone with or without bortezomib in previously untreated patients with MM who were ineligible for high-dose therapy. Patients (682 in total) were randomly assigned to receive 9 6-week cycles of melphalan (at a dose of 9 mg/m2 of body-surface area) and prednisone (at a dose of 60 mg/m2) on days 1–4, either alone or with bortezomib (at a dose of 1.3 mg/m2) on days 1, 4, 8, 11, 22, 25, 29, and 32 during cycles 1–4 and on days 1, 8, 22, and 29 during cycles 5–9. The primary end point was the time to disease progression. The time to progression among patients receiving bortezomib plus melphalan–prednisone (bortezomib group) was 24.0 months, as compared with 16.6 months among those receiving melphalan–prednisone alone (control group) (hazard ratio for the bortezomib group, 0.48; P < 0.001). Bortezomib plus melphalan–prednisone was superior to melphalan– prednisone alone in patients with newly diagnosed myeloma who were ineligible for high-dose therapy.
Another regimen designed to evaluate the effect of bortezomib as induction therapy before autologous SCT, used lenalidomide as consolidation maintenance in patients with MM.32 Newly diagnosed patients aged 65–75 years were eligible and induction (PAD) included 4 21-day cycles of bortezomib (1.3 mg/m2 on days 1, 4, 8, and 11), pegylated liposomal doxorubicin (30 mg/m2 on day 4), and dexamethasone (40 mg/d; cycle 1: days 1–4, 8–11, and 15–18; cycles 2–4: days 1–4). Autologous transplantation was tandem melphalan 100 mg/m2 (MEL100) and stem-cell support. Consolidation included 4 28-day cycles of lenalidomide (25 mg/d on days 1–21 every 28 days) + prednisone (50 mg every other day), followed by maintenance with lenalidomide (LP-L; 10 mg/d on days 1–21) until relapse. Bortezomib as induction before autologous transplantation, followed by lenalidomide as consolidation maintenance, was shown to be an effective regimen.
Bortezomib plus melphalan and prednisone (VMP) is significantly better than melphalan + prednisone alone for elderly patients with untreated MM; however, toxic effects are high.52 This combination was investigated as a novel and less intensive bortezomib-based regimen to maintain efficacy and to reduce toxic effects. MM patients (260) with untreated disease, 65 years and older, from 63 Spanish centers, were randomly assigned to receive 6 cycles of VMP (n = 130) or bortezomib plus thalidomide and prednisone (VTP; n = 130) as induction therapy. Induction consisted of 1 cycle of bortezomib twice per week for 6 weeks (1.3 mg/m2 on days 1, 4, 8, 11, 22, 25, 29, and 32), plus either melphalan (9 mg/m2 on days 1–4) or daily thalidomide (100 mg), and prednisone (60 mg/m2 on days 1–4). The first cycle was followed by 5 cycles of bortezomib once a week for 5 weeks (1.3 mg/m2 on days 1, 8, 15, and 22) plus the same doses of melphalan plus prednisone and thalidomide plus prednisone. A total of 178 patients completed the 6 induction cycles and were randomly assigned to maintenance therapy (MT) with bortezomib + prednisone (n = 87) or bortezomib + thalidomide (n = 91) that consisted of 1 conventional cycle of bortezomib for 3 weeks (1.3 mg/m2 on days 1, 4, 8, and 11) every 3 months, plus either prednisone (50 mg every other day) or thalidomide (50 mg/d), for up to 3 years. Treatment codes were generated with a computerized random number generator, and neither participants nor study personnel were masked to treatment. The primary endpoint was RR in induction and maintenance phases. Reduced-intensity induction with a bortezomib-based regimen, followed by maintenance, is a safe and effective treatment for elderly patients with multiple myeloma.
Bortezomib has also been investigated as an agent to improve clinical response in the postallogeneic SCT setting. The preclinical hypothesis of this use involves this drug blocking the nuclear factor-κB pathway that has been reported to exhibit multiple affects that include enhanced myeloma cell proliferation, a role in the inflammatory response pathway, enhanced apoptosis of alloreactive T cells and to promote cytotoxic T lymphocyte–induced tumor cell death to thus improve the graft-vs-myeloma response. Bruno et al36,53 treated 23 MM patients with relapsed disease postallograft with bortezomib alone in varying doses and schedules or in combination with corticosteroids. The ORR was 61% including 22% CR rate, median PFS of 6 months, 91% OS at median of 6 months after treatment with bortezomib with 7 still in ≥VGPR. Yet, no significant increase in graft-versus-host disease (GVHD) was reported. Kroger et al37 examined the use of bortezomib after reduced-intensity conditioning allo-SCT in 18 patients who achieved at least SD. Bortezomib 1.3 mg/m2 IV was administered on days 1, 4, 8, and 11 every 21 days for 2 cycles at a median of 8 months after allo-SCT. Ten patients had measurable disease at the time of bortezomib administration, and 3 patients achieved CR, 5 patients achieved PR, and 2 patients had an magnetic resonance imaging. There was a significant increase of neuropathy in patients also on cyclosporine: 3 patients had a mild aggravation of skin GVHD, and 1 patient developed grade I acute GVHD. It is noteworthy that 2 other studies done showed improvement in patients who had myeloma relapse after allo-SCT and had chronic GVHD.54 Follow-up study from Kröger et al demonstrated efficacy in the combination of bortezomib (in 8 of 32 patients) along with other agents (thalidomide and lenalidomide) in conjunction with donor lymphocyte infusion. This approach increased responses from PR to CR and significantly improved 5-year PFS.
MAINTENANCE THERAPY
Several studies have demonstrated its efficacy as frontline therapy and in relapsed, advanced MM, but results as MT in refractory/relapsed MM are less abundant.54 The combination bortezomib/dexamethasone can be safely administered as an MT in relapse/refractory MM. These preliminary data suggest that bortezomib/dexametasone MT combinations improve remission duration with acceptable toxicity. In newly diagnosed MM, the combination of VMP was superior to MP.51 In relapsed/refractory MM patients, the 4 drug combination bortezomib-melphalan-prednisone-thalidomide (VMPT) induced a relatively high proportion of CRs. The study was a prospective, randomized, phase III trial, compared VMPT with a maintenance regimen including bortezomib and thalidomide with VMP without a maintenance regimen and PFS as the primary end point. Weekly infusion of bortezomib reduced the incidence of peripheral neuropathy without affecting outcome with VMPT followed by maintenance with bortezomib and thalidomide superior to VMP for RRs and PFS. This was the first report to show the superiority of a 4-drug combination followed by maintenance.
CONCLUSIONS
In the field of cancer biology, the clinical success of the proteasome inhibitor bortezomib rapidly propelled the hematologic malignancy MM to a position of prominence as an efficient, validated preclinical model system to identify genetic lesions that contribute to disease, to unravel the pathways that contribute to oncogenesis and to test the therapeutic efficacy of novel compounds. Moreover, the availability of abundant, highly purified tumor samples from newly diagnosed, untreated patients has provided a platform for gene expression profiling to further identify genes deregulated during myelomagenesis in both selected and selected patient populations and has further accelerated the ascent of the MM field to the forefront of the cancer biology arena.55–57 Several therapeutic agents, such as the next generation proteasome inhibitors, now in clinical development were initially tested in myeloma systems. Seminal contributions from the basic biology and preclinical areas, such as linking the proteasome to the Ubiquitin-dependent proteolytic pathway, have translated into the clinical success of proteasome-based therapy in MM.58,59
The clinical success of bortezomib in the treatment of relapsed/refractory and newly diagnosed MM has led to novel combinations and applications as monotherapy, induction and MT. These therapies will be further refined to maximize benefit with minimal toxicity. Moreover, clinical trials with bortezomib as monotherapy have provided a basis for novel combinations such as combination chemotherapy regimen using lenalidomide, bortezomib, and dexamethasone in newly diagnosed myeloma patients.44 An unprecedented 100% of patients treated at the defined phase 2 dose level responded to treatment: 74% of patients experienced a 90% reduction in tumor burden and 57% entered a CR within a few months of starting treatment. As noted, by Stewart,60 there is little doubt that lenalidomide, bortezomib, and dexamethasone represents a step forward in myeloma care and serves as an important platform on which to build. Emerging proteasome inhibitors such as carfilzomib, NPI-0024, MLN9708, and CEP-18770, may demonstrate efficacy in additional malignancies and may overcome resistance to bortezomib that inevitably develops through unidentified means.
In recent years, it has become increasingly apparent that deregulation of the Ubiquitin + proteasome pathway contributes to the development of many forms of human cancer.61 Ubiquitin and many ubiquitin-like modifiers, for example, Small Ubiquitin-like MOdifier (SUMO) and NEDD8, are crucial signals that control many biological processes and function in pathologies to represent an important class of targets for human therapeutics. Current efforts are focused on understanding how these ubiquitin-linked pathways participate in the etiology of different tumor types.62–65 The SUMOylation pathway has been shown to be induced during myelomagenesis, to promote myeloma cell growth and proliferation and drug-resistance. Moreover, a gene signature comprising SUMO + Ub + Proteasome pathway components has been identified in patients with MM to predict response to bortezomib-based therapy. Targeting components of the Ubiquitin-like SUMOylation and NEDDylation pathways in malignancies may lead to promising tools as forms of cancer therapy.
Footnotes
The views expressed in this article are those of the authors and not official views of the Department of Defense.
The authors have no conflicts of interest to declare.
REFERENCES
- 1.Kyle RA, Rajkumar SV. Multiple Myeloma. NEJM. 2004;351:1860–1873. doi: 10.1056/NEJMra041875. [DOI] [PubMed] [Google Scholar]
- 2.Rajkumar SV, Kyle RA. Plasma cell disorders. In: Goldman L, Ausiello D, editors. Cecil Textbook of Medicine. 23rd ed. Philadelphia, PA: Saunders; 2007. pp. 1426–1437. [Google Scholar]
- 3.San Miguel J, Bladé J, Boccadoro M, et al. A practical update on the use of bortezomib in the management of multiple myeloma. Oncologist. 2006;11:51–61. doi: 10.1634/theoncologist.11-1-51. [DOI] [PubMed] [Google Scholar]
- 4.Strobeck M. Multiple myeloma therapies. Nat Rev Drug Discovery. 2007;6:181–182. doi: 10.1038/nrd2269. [DOI] [PubMed] [Google Scholar]
- 5.Rajkumar SV, Blood E, Vesole D, et al. Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the eastern cooperative oncology group. J Clin Oncol. 2006;24:431–436. doi: 10.1200/JCO.2005.03.0221. [DOI] [PubMed] [Google Scholar]
- 6.Myeloma trialists’ collaborative group: Combination chemotherapy versus melphalan plus prednisone as treatment for multiple myeloma: an overview of 6.633 patients from 27 randomized trials. J Clin Oncol. 1998;16:3832–3842. doi: 10.1200/JCO.1998.16.12.3832. [DOI] [PubMed] [Google Scholar]
- 7.Attal M, Harousseau JL, Stoppa AM, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma: Intergroupe Francais du Myelome. New Eng J Med. 1996;335:91–97. doi: 10.1056/NEJM199607113350204. [DOI] [PubMed] [Google Scholar]
- 8.Child JA, Morgan GJ, Davies FE, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Eng J Med. 2003;348:1875–1883. doi: 10.1056/NEJMoa022340. [DOI] [PubMed] [Google Scholar]
- 9.Boccadoro M, Palumbo A, Bringhen S, et al. Oral melphalan at diagnosis hampers adequate collection of peripheral blood progenitor cells in multiple myeloma. Haematologica. 2002;87:846–850. [PubMed] [Google Scholar]
- 10.Shaughnessy J, Jr, Tian E, Sawyer J, et al. Prognostic impact of cytogenetic and interphase fluorescence in situ hybridization-defined chromosome 13 deletion in multiple myeloma: early results of total therapy II. Br J Haematol. 2003;120:44–52. doi: 10.1046/j.1365-2141.2003.03948.x. [DOI] [PubMed] [Google Scholar]
- 11.Greipp PR, San Miguel J, Durie BG, et al. International staging system for multiple myeloma. J Clin Oncol. 2005;23:3412–3420. doi: 10.1200/JCO.2005.04.242. [DOI] [PubMed] [Google Scholar]
- 12.Avet-Loiseau H, Attal M, Moreau P, et al. Genetic abnormalities and survival in multiple myeloma: the experience of the Intergroupe Francophone du Myelome. Blood. 2007;109:3489–3495. doi: 10.1182/blood-2006-08-040410. [DOI] [PubMed] [Google Scholar]
- 13.Hideshima T, Mitsiades C, Akiyama M, et al. Molecular mechanisms mediating antimyeloma activity of proteasome inhibitor PS-341. Blood. 2003;101:1530–1534. doi: 10.1182/blood-2002-08-2543. [DOI] [PubMed] [Google Scholar]
- 14.Adams J, Palombella VJ, Sausville EA, et al. Proteasome inhibitors: a novel class of potent and effective antitumor agents. Cancer Res. 1999;59:2615–2622. [PubMed] [Google Scholar]
- 15.Wright JJ. Combination therapy of bortezomib with novel targeted agents: an emerging treatment strategy. Clin Cancer Res. 2010;16:4094–4104. doi: 10.1158/1078-0432.CCR-09-2882. [DOI] [PubMed] [Google Scholar]
- 16.Jagannath S, Barlogie B, Berenson J, et al. CREST Investigators. A phase II multicenter randomized study of the proteasome inhibitor bortezomib (VELCADE™ formerly PS-341) in multiple myeloma patients relapsed after front-line therapy [abstract] Blood. 2002;100:3027. [Google Scholar]
- 17.Richardson PG, Barlogie B, Berenson J, et al. SUMMIT Investigators. A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med. 2003;348:2609–2617. doi: 10.1056/NEJMoa030288. [DOI] [PubMed] [Google Scholar]
- 18.Richardson PG, Sonneveld P, Schuster MW, et al. APEX investigators. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Eng J Med. 2005;352:2487–2498. doi: 10.1056/NEJMoa043445. [DOI] [PubMed] [Google Scholar]
- 19.Fisher RI, Bernstein SH, Kahl BS, et al. PINNACLE Investigators. Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma. J Clin Oncol. 2006;24:4867–4874. doi: 10.1200/JCO.2006.07.9665. [DOI] [PubMed] [Google Scholar]
- 20.San Miguel JF, Schlag R, Khuageva NK, et al. VISTA Investigators. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Eng J Med. 2008;359:906–917. doi: 10.1056/NEJMoa0801479. [DOI] [PubMed] [Google Scholar]
- 21.Driscoll JJ, DecHowdhury R, Burriss J, et al. The expanding role of proteasome-based therapy in the treatment of hematologic malignancies. Open J Hematology. 2010;1:1–20. [Google Scholar]
- 22.Dispenzieri A, Jacobus S, Vesole DH, et al. Primary therapy with single agent bortezomib as induction, maintenance and re-induction in patients with high-risk myeloma: results of the ECOG E2A02 trial. Leukemia. 2010;24:1406–1411. doi: 10.1038/leu.2010.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bladé J, Rosiñol L, Cibeira MT, et al. Hematopoietic stem cell transplantation for multiple myeloma beyond 2010. Blood. 2010;115:3655–3663. doi: 10.1182/blood-2009-08-238196. [DOI] [PubMed] [Google Scholar]
- 24.Barlogie B, Anaissie E, van Rhee F, et al. Incorporating bortezomib into upfront treatment for multiple myeloma: early results of total therapy 3. Br J Haematol. 2007;138:176–185. doi: 10.1111/j.1365-2141.2007.06639.x. [DOI] [PubMed] [Google Scholar]
- 25.Nair B, van Rhee F, Shaughnessy JD, Jr, et al. Superior results of total therapy 3 (2003–33) in gene expression profiling–defined low-risk multiple myeloma confirmed in subsequent trial 2006–66 with VRD maintenance. Blood. 2010;115:4168–4173. doi: 10.1182/blood-2009-11-255620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jagannath S, Durie BG, Wolf J, et al. Bortezomib therapy alone and in combination with dexamethasone for previously untreated symptomatic multiple myeloma. Br J Haematol. 2005;129:776–783. doi: 10.1111/j.1365-2141.2005.05540.x. [DOI] [PubMed] [Google Scholar]
- 27.Harousseau JL, Attal M, Leleu X, et al. Bortezomib plus dexamethasone as induction treatment prior to autologous stem cell transplantation in patients with newly diagnosed multiple myeloma: results of an IFM phase II study. Haematologica. 2006;91:1498–1505. [PubMed] [Google Scholar]
- 28.Rosinol L, Oriol A, Mateos MV, et al. Phase II PETHEMA trial of alternating bortezomib and dexamethasone as induction regimen before autologous stem-cell transplantation in younger patients with multiple myeloma: efficacy and clinical implications of tumor response kinetics. J Clin Oncol. 2007;25:4452–4458. doi: 10.1200/JCO.2007.12.3323. [DOI] [PubMed] [Google Scholar]
- 29.Richardson PG, Xie W, Mitsiades C, et al. Single-agent bortezomib in previously untreated multiple myeloma: efficacy, characterization of peripheral neuropathy, and molecular correlations with response and neuropathy. J Clin Oncol. 2009;27:3518–3525. doi: 10.1200/JCO.2008.18.3087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Popat R, Oakervee HE, Hallam S, et al. Bortezomib, doxorubicin and dexamethasone (PAD) frontline treatment of multiple myeloma: updated results after long-term follow-up. Br J Haematol. 2008;141:512–516. doi: 10.1111/j.1365-2141.2008.06997.x. [DOI] [PubMed] [Google Scholar]
- 31.Cavo M, Tacchetti P, Patriarca F, et al. A phase III study of double autotransplantation incorporating bor-tezomib/thalidomide/dexamethasone (VTD) or thalidomide/dexamethasone for multiple myeloma: superior clinical outcomes with VTD compared to TD [abstract] Blood (ASH Annual Meeting Abstracts) 2009;114:148. Abstract 351. [Google Scholar]
- 32.Palumbo A, Gay F, Falco P, et al. Bortezomib as induction before autologous transplantation, followed by lenalidomide as consolidation-maintenance in untreated multiple myeloma patients. J Clin Oncol. 2010;28:800–807. doi: 10.1200/JCO.2009.22.7561. [DOI] [PubMed] [Google Scholar]
- 33.Moreau P, Facon T, Attal M, et al. Comparison of reduced-dose bortezomib plus thalidomide plus dexamethasone (vTD) to bortezomib plus dexamethasone (VD) as induction treatment prior to ASCT in de novo multiple myeloma (MM): results of IFM 2007-02 study. JCO. 2010;28 Abstract 8014. [Google Scholar]
- 34.Roussel M, Moreau P, Huynh J, et al. Bortezomib and high-dose melphalan as conditioning regimen before transplantation for de novo multiple myeloma patients: updated data of the IFM phase II study. JCO. 2010;28 Abstract 8129. [Google Scholar]
- 35.Harousseau JL, Attal M, Avet-Loiseau H, et al. Bortezomib plus dexamethasone is superior to vincristine plus doxorubicin plus dexamethasone as induction treatment prior to autologous stem-cell transplantation in newly diagnosed multiple myeloma: results of the IFM 2005-01 phase III trial. [Accessed September 7, 2010];JCO. doi: 10.1200/JCO.2009.27.9158. Available at: http://jco.ascopubs.org/cgi/doi/10.1200/JCO.2009.27.9158. [DOI] [PubMed] [Google Scholar]
- 36.Bruno B, Patriarca F, Sorasio R, et al. Bortezomib with or without dexamethasone in relapsed multiple myeloma following allogeneic hematopoietic cell transplantation. Haematologica. 2006;91:837–839. [PubMed] [Google Scholar]
- 37.Kroger N, Zabelina T, Ayuk F, et al. Bortezomib after dose-reduced allogeneic stem cell transplantation for multiple myeloma to enhance or maintain remission status. Exp Hematol. 2006;34:770–775. doi: 10.1016/j.exphem.2006.02.020. [DOI] [PubMed] [Google Scholar]
- 38.Kroger N, Badbaran A, Lioznov M, et al. Post-transplant immunotherapy with donor-lymphocyte infusion and novel agents to upgrade partial into complete and molecular remission in allografted patients with multiple myeloma. Exp Hematol. 2009;37:791–798. doi: 10.1016/j.exphem.2009.03.008. [DOI] [PubMed] [Google Scholar]
- 39.Sonneveld P, van der Holt B, Schmidt-Wolf IGH, et al. First Analysis of HOVON-65/GMMG-HD4: randomized phase III Trial comparing bortezomib, adriamycine, dexamethasone (PAD) vs VAD as induction treatment prior to high dose melphalan (HDM) in patients with newly diagnosed Multiple Myeloma (MM) Blood. (ASH Annual Meeting Abstracts) 2008;112 Abstract 653. [Google Scholar]
- 40.Rosinol L, Cibeira MT, Martinez J, et al. Thalidomide/dexamethasone (TD) Vs. bortezomib (Velcade®)/thalidomide/dexamethasone (VTD) vs. VBMCP/VBAD/Velcade® as induction regimens prior autologous stem cell transplantation (ASCT) in multiple myeloma (MM): results of a Phase III PETHEMA/GEM Tria. Blood. (ASH Annual Meeting Abstracts) 2009;114 Abstract 130. [Google Scholar]
- 41.Palumbo A, Bringhen S, Rossi D, et al. Bortezomib, melphalan, prednisone and thalidomide (VMPT) followed by maintenance with bortezomib and thalidomide for initial treatment of elderly multiple myeloma patients. Blood. (ASH Annual Meeting Abstracts) 2009;114 Abstract 128. [Google Scholar]
- 42.Ladetto M, Pagliano G, Ferrero S, et al. Major tumor shrinking and persistent molecular remissions after consolidation with bortezomib, thalidomide, and dexamethasone in patients with autografted myeloma. JCO. 2010;28:2077–2084. doi: 10.1200/JCO.2009.23.7172. [DOI] [PubMed] [Google Scholar]
- 43.Kumar SK, Flinn I, Noga SJ, et al. Bortezomib, dexamethasone, cyclophosphamide and lenalidomide combination for newly diagnosed multiple myeloma: phase 1 results from the multicenter EVOLUTION study. Leukemia. 2010;24:1350–1356. doi: 10.1038/leu.2010.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Richardson PG, Weller E, Lonial S, et al. Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma. Blood. 2010;116:679–686. doi: 10.1182/blood-2010-02-268862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Mateos MV, Oriol A, Martínez-López J, et al. Bortezomib, melphalan, and prednisone versus bortezomib, thalidomide, and prednisone as induction therapy followed by maintenance treatment with bortezomib and thalidomide versus bortezomib and prednisone in elderly patients with untreated multiple myeloma: a randomised trial. Lancet Oncol. 2010;11:934–941. doi: 10.1016/S1470-2045(10)70187-X. [DOI] [PubMed] [Google Scholar]
- 46.Attal M, Harousseau JL, Marit G, et al. Lenalidomide after autologous transplantation for myeloma: First analysis of a prospective randomized study of the Intergroupe Francophone Du Myelome (IFM 2005 02) Blood. (ASH Annual Meeting) 2009;14 abstract 529. [Google Scholar]
- 47.Cavo M, Tosi P, Zamagni E, et al. Prospective, randomized study of single compared with double autologous transplantation for multiple myeloma: Bologna 96 clinical study. J Clin. Oncol. 2007;25:2434–2441. doi: 10.1200/JCO.2006.10.2509. [DOI] [PubMed] [Google Scholar]
- 48.Orlowski RZ, Peterson BL, Sanford B, et al. Bortezomib and pegylated liposomal doxorubicin as induction therapy for adult patients with symptomatic multiple myeloma: cancer and leukemia group B study 10301. Blood. 2006;108:797. [Google Scholar]
- 49.Barlogie B, Haessler J, Pineda-Roman M, et al. Completion of premaintenance phases in total therapies 2 and 3 improves clinical outcomes in multiple myeloma: an important variable to be considered in clinical trial designs. Cancer. 2008;112:2720–2725. doi: 10.1002/cncr.23487. [DOI] [PubMed] [Google Scholar]
- 50.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 Lymphoma. 2007;48:1910–1921. doi: 10.1080/10428190701540991. [DOI] [PubMed] [Google Scholar]
- 51.San Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008;359:906–917. doi: 10.1056/NEJMoa0801479. [DOI] [PubMed] [Google Scholar]
- 52.Mateos M-V, Richardson PG, Schlag R, et al. Bortezomib plus melphalan-prednisone continues to demonstrate a survival benefit vs melphalan-prednisone in the Phase III VISTA Trial in previously untreated multiple myeloma after 3 years’ follow-up and extensive subsequent therapy use. ASH Ann Meet Abstr. 2009;114:3859. [Google Scholar]
- 53.Bruno B, Rotta M, Patriarca F, et al. Nonmyeloablative allografting for newly diagnosed multiple myeloma: the experience of the Gruppo Italiano Trapianti di Midollo. Blood. 2009;113:3375–3382. doi: 10.1182/blood-2008-07-167379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Benevolo G, Larocca A, Pregno P, et al. Bortezomib and dexamethasone as maintenance therapy in relapse/refractory multiple myeloma patients. Blood. (ASH Annual Meeting Abstracts) 2008;112:2771. [Google Scholar]
- 55.Zhan F, Hardin J, Kordsmeier B, et al. Global gene expression profiling of multiple myeloma, monoclonal gammopathy of undetermined significance, and normal bone marrow plasma cells. Blood. 2002;99:1745–1757. doi: 10.1182/blood.v99.5.1745. [DOI] [PubMed] [Google Scholar]
- 56.Shaughnessy J, Zhan F, Burington B, et al. A validated gene expression model of high-risk multiple myeloma is defined by deregulated expression of genes mapping to chromosome I. Blood. 2007;109:2276–2284. doi: 10.1182/blood-2006-07-038430. [DOI] [PubMed] [Google Scholar]
- 57.Burington B, Barlogie B, Zhan F, et al. Tumor cell gene expression changes following short-term in vivo exposure to single agent chemotherapeutics are related to survival in multiple myeloma. Clin Cancer Res. 2008;14:4821–4829. doi: 10.1158/1078-0432.CCR-07-4568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Eytan E, Ganoth D, Armon T, et al. ATP-dependent incorporation of 20S protease into the 26S complex that degrades proteins conjugated to ubiquitin. Proc Natl Acad Sci USA. 1989;86:7751–7755. doi: 10.1073/pnas.86.20.7751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Driscoll J, Goldberg AL. The proteasome (multicatalytic protease) is a component of the 1500-kDa proteolytic complex which degrades ubiquitin-conjugated proteins. J Biol Chem. 1990;265:4789–4792. [PubMed] [Google Scholar]
- 60.Stewart AK. Union of forces advances myeloma care. Blood. 2010:116674–116675. doi: 10.1182/blood-2010-04-281212. [DOI] [PubMed] [Google Scholar]
- 61.Hoeller D, Hecker CM, Dikic I. Ubiquitin and ubiquitin-like proteins in cancer pathogenesis. Nature Rev Cancer. 2006;6:776–788. doi: 10.1038/nrc1994. [DOI] [PubMed] [Google Scholar]
- 62.Soucy TA, Smith PG, Milhollen MA. An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature. 2009;458:732–736. doi: 10.1038/nature07884. [DOI] [PubMed] [Google Scholar]
- 63.Driscoll JJ, Pelluru D, Lefkimmiatis K, et al. The sumoylation pathway is dysregulated in multiple myeloma and is associated with adverse patient outcome. Blood. 2010;115:2827–2834. doi: 10.1182/blood-2009-03-211045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Milhollen MA, Traore T, Adams-Duffy J, et al. MLN4924, a NEDD8-activating enzyme inhibitor, is active in diffuse large B-cell lymphoma models: rationale for treatment of NF- κB–dependent lymphoma. Blood. 2010;116:1515–1523. doi: 10.1182/blood-2010-03-272567. [DOI] [PubMed] [Google Scholar]
- 65.Driscoll JJ, Amin S, Pelluru D. The poly-SUMOylation pathway confers resistance to proteasome inhibitor-based therapy of multiple myeloma. J Clin Invest. 2010 manuscript in preparation. [Google Scholar]