Proteasome inhibition has emerged as an important therapeutic strategy in multiple myeloma (MM). Since the publication of the first phase 1 trials of bortezomib 10 years ago, this first-in-class proteasome inhibitor (PI) has contributed substantially to the observed improvement in survival in MM patients over the past decade. Although first approved as a single agent in the relapsed setting, bortezomib is now predominantly used in combination regimens. Furthermore, the standard twice-weekly schedule may be replaced by weekly infusion, especially when bortezomib is used as part of combination regimens in frontline therapy. Indeed, bortezomib is an established component of induction therapy for patients eligible or ineligible for autologous stem cell transplantation. Bortezomib has also been incorporated into conditioning regimens before autologous stem cell transplantation, as well as into post-ASCT consolidation therapy, and in the maintenance setting. In addition, a new route of bortezomib administration, subcutaneous infusion, has recently been approved. Recently, several new agents have been introduced into the clinic, including carfilzomib, marizomib, and MLN9708, and trials investigating these “second-generation” PIs in patients with relapsed/refractory MMs have demonstrated positive results. This review provides an overview of the role of PIs in the treatment of MM, focusing on developments over the past decade.

Proteasome inhibition as a therapeutic strategy

The ubiquitin-proteasome pathway is responsible for degradation of the majority of regulatory proteins in eukaryotic cells, including proteins that control cell-cycle progression, apoptosis, and DNA repair and therefore plays an essential role in maintaining normal cellular homeostasis.1-3  The 26S proteasome consists of a barrel-shaped 20S proteolytic core, composed of 2 identical α-subunit rings and 2 identical β-subunit rings, plus 2 19S regulatory complexes that cap the 20S barrel.4  Proteins destined for degradation are first polyubiquitinated; the 19S cap recognizes and binds ubiquitinated proteins and directs them to the 20S core, where proteolytic cleavage is mediated by 3 β-subunits: β1 (caspase-like activity), β2 (trypsin-like activity), and β5 (chymotrypsin-like activity).1-3  Disruption of proteasome activity results in growth arrest and cell death because of induction of an apoptotic cascade as a result of the rapid accumulation of incompatible regulatory proteins within the cell.5 

Cancer cells generally have higher levels of proteasome activity compared with normal cells and, moreover, are more sensitive to the proapoptotic effects of proteasome inhibition than normal cells,6  making the proteasome a rational therapeutic target in oncology.7  Based on promising preclinical results, proteasome inhibition has been extensively explored as a therapeutic strategy in multiple myeloma (MM), and proteasome inhibitors (PIs) now form a cornerstone of antimyeloma therapy.

Bortezomib was the first-in-class PI to be introduced into the clinic. Recently, a number of second-generation PIs have been developed and are undergoing intense examination in clinical trials. The different PIs are distinct regarding their specificities and affinities for the different catalytic sites within the proteasome core (Table 1).8-10  Based on chemical structure and active moiety, PIs can be classified into 3 groups: boronates, epoxyketones, and salinosporamides.

Boronates.

Bortezomib is a dipeptidyl boronic acid-based specific, reversible PI that targets the chymotrypsin- and caspase-like active sites, with minimal effect on trypsin-like activity.8  By inhibiting the proteasome, bortezomib acts through multiple mechanisms to suppress tumor survival pathways and to arrest tumor growth, tumor spread, and angiogenesis.11  The mechanisms of its antitumor activity in MM have been elucidated through in vitro and in vivo experiments. Bortezomib directly induces apoptosis of tumor cells, inhibits the activation of NF-κB in cells and in the tumor microenvironment, reduces adherence of myeloma cells to bone marrow stromal cells, blocks production and intracellular signaling of IL-6 in myeloma cells, stops the production and expression of proangiogenic mediators, and overcomes defects in apoptotic regulators, such as Bcl-2 overexpression and alterations in tumor suppressor p53.11,12  In addition, proteasome inhibition with bortezomib has been shown to induce the endoplasmic reticulum stress response, associated with disruption of the unfolded protein response, an important aspect of the mechanism of action of proteasome inhibition in MM because of the high production of immunoglobulins by MM cells.13  These mechanisms of action of proteasome inhibition have provided the rationale for the combination of bortezomib and other PIs with numerous chemotherapeutic and targeted agents12-14 ; for example, the disruption of protein quality control and the consequent activation of aggresomal degradation of misfolded proteins have provided a strong rationale for the combination of PIs and histone deacetylase inhibitors.13  As a result of its mechanisms of action, bortezomib has also been associated with increased bone formation and osteoblastic activity, and decreased bone resorption and osteoclastic activity.15 

MLN9708 is another boronate PI that is a reversible inhibitor of primarily the chymotrypsin-like activity of the 20S proteasome. However, in contrast to bortezomib, MLN9708 has a shorter dissociation half-life (Table 1) and has demonstrated greater tissue penetration compared with bortezomib in preclinical studies.10  Furthermore, MLN9708 is orally available and is the first oral PI to enter clinical trials in MM. Like bortezomib, MLN9708 has been shown to inhibit NF-κB activation and demonstrated antitumor activity in MM and other hematologic malignancies in in vitro and in vivo studies.10  A third boronate PI, CE P-18770,16  was also in development, but data from clinical trials have not been reported.

Epoxyketones.

Carfilzomib (PR-171) is an irreversible PI that belongs to the epoxyketone class and is structurally and mechanistically distinct from bortezomib.17  It demonstrates potent and sustained inhibition of chymotrypsin-like activity and appears to have a greater selectivity for the chymotrypsin-like protease compared with bortezomib, with lower affinity for the trypsin- and caspase-like proteases.9  Unlike the boronate PIs, carfilzomib has minimal activity against off-target enzymes, including serine proteases.18  Like bortezomib, carfilzomib inhibits both the constitutive and immunoproteasome, and carfilzomib has equivalent potency against the β5 and LMP7 subunits (Table 1).19 

Carfilzomib has been shown to trigger cell cycle arrest, induce apoptosis, and activate stress response pathways in human tumor cell lines, including MM and other hematologic malignancies and solid tumors.17  Importantly, carfilzomib has demonstrated activity against bortezomib-resistant cell lines and primary MM cells.19  The carfilzomib analog ONX 0912 is another peptide epoxyketone PI that is in development20 ; however, no data from clinical studies of this orally available agent have yet been reported. In addition to antimyeloma effects, these epoxyketone PIs have also been shown to inhibit bone resorption in preclinical models.21 

Salinosporamides.

Marizomib (NPI-0052) is a natural lactone compound derived from the marine bacterium Salinospora tropica. This agent belongs to a unique class of PIs, the salinosporamides. Marizomib is an irreversible PI that, unlike bortezomib and carfilzomib, inhibits both the chymotrypsin-like and trypsin-like protease activities, but only minimally affects the caspase-like activity within the proteasome. As a result, marizomib has a unique efficacy and safety profile and does not exhibit cross-resistance with other PIs. Marizomib-induced apoptosis is predominantly the result of activation of caspase-8–mediated signaling pathways.22  Marizomib has demonstrated antitumor activity in preclinical models of MM, other hematologic malignancies, and solid tumors.23 

Establishing efficacy and safety: initial studies of single-agent bortezomib

The efficacy and safety of bortezomib were established through a number of important phase 1 to 3 studies. The earliest phase 1 studies, which were reported 10 years ago, demonstrated manageable toxicity with promising antineoplastic activity, particularly in patients with relapsed/refractory MM.24-26  When administered intravenously (IV) on a twice-weekly schedule, the maximum tolerated dose (MTD) was determined to be 1.3 mg/m2, based on dose-limiting adverse events (AEs) of diarrhea and peripheral neuropathy (PN).24  Notable in these phase 1 studies was the relative lack of significant myelosuppression; in addition, cardiac, hepatic, or renal toxicities were infrequent.

After these early studies, the efficacy of bortezomib in MM was confirmed in the large phase 2 SUMMIT study (Table 2), which investigated bortezomib in relapsed/refractory MM patients.27  Overall, 27% of patients achieved partial response (PR) or better, including 10% complete/near-complete responses (CR/nCR). Median time to progression (TTP) was 7 months, compared with 3 months with patients' previous therapy. Another phase 2 study (CREST) prospectively compared 2 doses of bortezomib in patients with relapsed or refractory MM.28  Of note, the survival, response, and TTP data suggested that a starting dose of 1.3 mg/m2 was preferable; however, the trial also showed that the dose of bortezomib could be reduced to 1.0 mg/m2 if required while still offering patients a substantial survival benefit, thereby providing valuable information regarding the use of this agent in clinical practice.29  On the basis of these phase 2 results, bortezomib was approved for the treatment of relapsed and refractory MM and moved into phase 3 investigation. The international phase 3 APEX trial demonstrated the superiority of bortezomib over high-dose dexamethasone,30  which at that time could be considered a standard of care in the relapsed setting. Bortezomib demonstrated superior response rates, TTP (the primary endpoint), and overall survival (OS; Table 2),30  and, in an updated analysis with a median follow-up of 22 months, median OS was 29.8 months versus 23.7 months, despite more than 62% of dexamethasone patients crossing over to receive bortezomib.31  Based on these results, bortezomib soon became one of the treatments of choice for relapsed MM.

These initial studies of single-agent bortezomib also established the common bortezomib-associated toxicities, the most frequent being gastrointestinal symptoms, anemia, thrombocytopenia, fatigue, and PN. As treatment-related PN was noted in these key studies, substantial efforts were made to characterize this toxicity. It is thought that bortezomib mainly causes direct dorsal root ganglion toxicity, based on studies in mice, which showed that ubiquitinated aggregates accumulated in the cytoplasm of the dorsal root ganglion.32-34  There is now substantial experience regarding bortezomib-induced PN, and a number of excellent reviews provide a comprehensive overview of its etiology, clinical characteristics, and management.35-38  Bortezomib-induced PN can be effectively managed with dose modification and is generally reversible in more than 50% of cases, with weekly dosing39-41  and subcutaneous administration of bortezomib providing approaches to limit treatment-emergent PN, as described in more detail later.42  The most common hematologic toxicity associated with bortezomib is transient thrombocytopenia,43  which shows a cyclical pattern of platelet decrease and recovery, without evidence of cumulative thrombocytopenia.43,44  The transient, predictable decrease in platelet counts generally results in a low requirement for platelet support, and so bortezomib can usually be administered to thrombocytopenic patients.44 

Subcutaneous administration of bortezomib

As an alternative to the previous standard of IV administration, bortezomib has been infused subcutaneously, and this route of administration has recently been approved by the United States Food and Drug Administration. A randomized phase 1 trial of subcutaneous (SC) versus IV bortezomib in relapsed/refractory MM showed similar systemic bortezomib exposure, 20S proteasome inhibition, response rates, and safety with SC and IV bortezomib.45  A large phase 3 study has confirmed these preliminary results (Table 2),42  demonstrating the noninferiority of SC versus IV bortezomib in terms of overall response rate (ORR) after 4 cycles. After median follow-up of 12 months, there were no significant differences in TTP and 1-year OS with SC versus IV bortezomib. PN of any grade (38% vs 53%; P = .04), grade more than or equal to 2 (24% vs 41%; P = .01), and grade more than or equal to 3 (6% vs 16%; P = 0·03) was significantly less common with SC than IV administration. These results have validated a more convenient and less toxic route of administration that is likely to become standard in the near future.

Re-treatment with bortezomib for relapsed MM

Given the finite treatment course of bortezomib, as used in the phase 2 and 3 studies described earlier, patients may remain sensitive to bortezomib after relapse after initial therapy. Studies have specifically addressed the issue of bortezomib retreatment in relapsed MM, confirming that it is feasible, without evidence of cumulative toxicity. In a retrospective multicenter survey of 94 relapsed MM patients who had responded to initial bortezomib treatment, no uncommon toxicity with bortezomib retreatment was identified, and efficacy data showed that 63% of patients responded to retreatment, with a median TTP of 9.3 months, consistent with sustained sensitivity to bortezomib.46  A similar prospective phase 2 trial demonstrated an ORR of 32% and 42% in patients who received single-agent bortezomib and bortezomib-dexamethasone, respectively, as retreatment after initial response to bortezomib.47 

Bortezomib-based combinations in relapsed/refractory MM

Subsequent to the demonstration of single-agent activity, studies showed that the efficacy of bortezomib was enhanced through combination with agents with different modes of action. Based on observations from in vitro studies of synergistic activity between bortezomib and anthracyclines,48  and on phase 1 data,49  a phase 3 study compared single-agent bortezomib with bortezomib plus pegylated liposomal doxorubicin (PLD) in patients with relapsed/refractory MM (Table 2).50  The combination was superior, with median TTP increased from 6.5 to 9.3 months with bortezomib-PLD versus bortezomib, albeit at the cost of increased grade 3 or 4 toxicity. These results led to the approval of the combination for relapsed MM in 2007. In vitro studies also showed that dexamethasone and bortezomib act synergistically,51  and clinical observations showed that the addition of dexamethasone improved responses in up to 34% of patients after suboptimal response to single-agent bortezomib.42,52,53  Although no randomized trial comparing bortezomib versus bortezomib-dexamethasone has been performed, it is now common practice to add dexamethasone, with or without other agents, to bortezomib when treating relapsed MM.

Apart from the bortezomib-PLD trial, few data on bortezomib-based combinations in the relapsed setting are available from randomized phase 3 trials. Recently, the Intergroupe Francophone du Myélome (IFM) and European Group for Blood and Marrow Transplantation conducted a prospective comparison of bortezomib-thalidomide-dexamethasone (VTD) versus thalidomide-dexamethasone (TD) in patients relapsing after high-dose therapy plus autologous stem cell transplantation (HDT-ASCT) and showed that median TTP was significantly longer with VTD than TD, with a trend for a survival benefit (Table 2).54  This study showed, for the first time, that a triplet bortezomib-based combination was superior to a 2-drug thalidomide-based regimen for relapsed MM, in terms of significant greater response rates and longer TTP. Less striking, the VANTAGE 088 phase 3 trial showed only a weak clinical benefit with bortezomib plus the histone deacetylase-inhibitor vorinostat versus single-agent bortezomib55 ; median progression-free survival (PFS) benefit was just 0.8 months, indicating that alternative combinations should be tested. Currently, other novel agents, such as perifosine or panobinostat, are being evaluated in combination with bortezomib in phase 3 trials.

Multiple other bortezomib-based combinations have been studied in phase 1 or 2 trials, incorporating steroids plus alkylators, immunomodulatory drugs (IMiDs), monoclonal antibodies, and heat shock protein-90, histone deacetylase, or AKT inhibitors.14,56  Widely used regimens include bortezomib plus cyclophosphamide and dexamethasone (VCD) or prednisone (VCP), or bortezomib plus lenalidomide and dexamethasone (VRD). Response rates range from 60% to 90%, with median PFS of approximately 7 to 15 months and median OS of 16 to 37 months, and bortezomib can be delivered at or near the standard dose and schedule with few overlapping toxicities.

Bortezomib in frontline MM

VMP-based combinations for transplant-ineligible patients.

For many years, the standard of care in elderly patients was melphalan-prednisone (MP). Preclinical studies demonstrated in vitro synergy with bortezomib combined with melphalan,48  and consequently bortezomib was studied in combination with MP (VMP). A phase 1 or 2 trial of VMP demonstrated an ORR of 89%, including 32% CR,57  median TTP of 27 months, and OS at 38 months of 85%.58  These impressive results were the basis for the randomized phase 3 VISTA trial that compared MP versus VMP in previously untreated MM patients ineligible for HDT-ASCT (Table 3).59  At the first report, median TTP (primary endpoint) was 24.0 versus 16.6 months with VMP versus MP (P < .001),59  the ORR was 71% versus 35%, and CR rates were 30% and 4%, respectively (P < .001). Importantly, the hazard ratio for OS was 0.61 in favor of VMP, with a median follow-up of 16.3 months.59  These results led to the approval of VMP as induction therapy in patients ineligible for ASCT in the United States and European Union, and this regimen is now considered one of the standards of care in this population. Updated results confirming a survival advantage for VMP have been reported,60,61  with 5-year follow-up data demonstrating a continued statistically significant OS benefit with VMP versus MP despite a higher proportion of MP patients receiving subsequent bortezomib (43% vs 22%).61 

In both these VMP studies,57,59  important toxic effects were recorded, particularly PN and gastrointestinal symptoms. Consequently, the Spanish group designed a novel, less intensive bortezomib-based regimen to maintain efficacy while reducing toxic effects (Table 3).40  Patients received induction therapy based mainly on weekly rather than twice-weekly dosing of bortezomib, with prednisone plus melphalan (VMP) or thalidomide (VTP). This was followed by a second randomization to maintenance with bortezomib-thalidomide (VT) or bortezomib-prednisone (VP). There was no difference in response rates after induction between VMP and VTP, but the grade 3 or 4 PN rate was only 7% with VMP arm (half that in VISTA). After maintenance, the CR rate was approximately 40% in both arms, and only 3% of VP and 9% of VT patients developed grade 3 PN.62  Overall median PFS and TTP were 31 and 35 months, respectively, and the 3-year OS was 70%, which compared favorably with VISTA results. Therefore, the goal of reducing toxicity without impairing outcomes was achieved.

Another randomized phase 3 trial has provided data on weekly administration of bortezomib and the potential of bortezomib-based maintenance. The Italian group examined VMP-thalidomide (VMPT) followed by VT maintenance (VMPT-VT) compared with VMP alone in transplant-ineligible patients (Table 3).39,41  comparing the 2 arms of the study, the efficacy results (including response rates and PFS) were in favor of VMPT-VT. After inclusion of the first 139 patients, the protocol was amended with the aim of reducing the incidence of PN, and both VMPT-VT and VMP induction schedules were changed from the planned VISTA-like twice-weekly schedule to compose nine 5-week cycles using weekly bortezomib dosing. Remarkably, there was a substantial reduction in the incidence of nonhematologic severe AEs in the weekly versus twice-weekly group, whereas long-term outcomes appeared similar (Table 3).39  Notably, the incidence of any grade 3 or 4 PN was 8% versus 28% with weekly versus twice-weekly bortezomib (P < .001). The authors concluded that the weekly schedule resulted in a substantial improvement in safety and did not impact efficacy.

These 2 phase 3 trials are of major importance because they validate the weekly VMP schedule in elderly patients, demonstrating that this modified schedule is able to significantly decrease toxicity, especially PN, without adversely affecting the overall outcome. The studies, together with the phase 3b community-based UPFRONT study (Table 3), have also introduced the concept of bortezomib-based maintenance in elderly patients. To date, data regarding a role for bortezomib maintenance in this setting remain inconclusive because the Spanish trial proposed maintenance in both arms and the Italian trial used different induction therapy in the 2 arms. Data from a “pure” maintenance trial comparing bortezomib versus no bortezomib after a common induction therapy are currently not available.

Bortezomib-based induction before ASCT.

The activity of bortezomib in the relapsed setting prompted its evaluation upfront as part of induction before HDT-ASCT. Several phase 2 trials investigated bortezomib-dexamethasone induction, with response rates of 66% to 90%, including 15% to 21% CR and 31% to 70% very good PR (VGPR) or better.63-65  This translated into high CR and VGPR rates after transplantation. Toxicities were generally mild to moderate and proved manageable, without treatment-related mortality, and stem cell collection was adequate. Therefore, this promising combination was compared prospectively with vincristine-doxorubicin-dexamethasone (VAD), which was considered the standard induction regimen.

The IFM2005-01 phase 3 study compared bortezomib-dexamethasone with VAD as pre-ASCT induction in previously untreated patients (Table 3).66  Postinduction response rates were significantly higher with bortezomib-dexamethasone, regardless of disease stage or adverse cytogenetic abnormalities. After first transplantation, CR/nCR and more than or equal to VGPR rates remained significantly higher with bortezomib-dexamethasone (Table 3). There was a trend for improved PFS with bortezomib-dexamethasone versus VAD (P = .06), but 3-year survival rates were not different. It should be noted that more than half the patients in each group subsequently received lenalidomide consolidation and were randomized to lenalidomide or placebo maintenance as part of another IFM study. The incidence of severe AEs appeared similar between groups, but hematologic toxicity and treatment-related mortality were more frequent with VAD. Conversely, rates of grade 3 or 4 PN during induction through first transplantation were significantly higher with bortezomib-dexamethasone than VAD (9.2% vs 2.5%). The level of response achieved with bortezomib-dexamethasone after induction in IFM2005-01 is now considered the goal for current therapies; as a result, bortezomib-dexamethasone has become the backbone of pre-ASCT induction therapy to which other, more complex regimens should be compared.

The addition of a third agent to bortezomib-dexamethasone, including thalidomide (VTD),67  (liposomal) doxorubicin (VDD/PAD),68,69  lenalidomide (VRD),70  and cyclophosphamide (VCD),71  has been tested in several phase 2 studies, and outcomes appear even better than with the doublet. Three prospective studies have already shown that VTD is superior to TD or bortezomib-dexamethasone in terms of response rates (Table 3).72-74  The Italian group compared TD versus VTD as induction before, and consolidation after, tandem ASCT and found that VTD resulted in higher CR and more than or equal to VGPR rates after induction and after ASCT, which translated into better PFS (Table 3).72  The Spanish group also compared TD versus VTD versus a more complex chemotherapy regimen, including bortezomib, before ASCT, and confirmed that VTD achieved the best pre- and post-ASCT CR rates.74  In the IFM2007-02 trial, 4 cycles of “standard” bortezomib-dexamethasone induction were compared with 4 cycles of VTD, using lower bortezomib and thalidomide doses to reduce the neuropathy rate.73  VTD was again found to result in superior response rates before and after ASCT, and the reduced bortezomib and thalidomide doses were associated with reduced neurotoxicity. The IFM2007-02 study therefore provided further evidence for the superiority of a 3-drug over a 2-drug combination for tumor burden reduction as pre-ASCT induction. Further, the HOVON group reported a phase 3 randomized trial comparing VAD versus PAD as pre-ASCT induction, followed by thalidomide and bortezomib maintenance, respectively.75  This study confirmed the superiority of the bortezomib-based triplet over VAD in terms of postinduction and post-ASCT response rates; and, notably, OS was also superior after PAD plus bortezomib maintenance.

No data are available to draw conclusions regarding the superiority of one bortezomib-based combination over another. Although response rates are clearly improved with novel agent-based regimens, demonstration of a significant OS advantage will often be difficult given the large numbers of patients and the long duration of follow-up required, and the availability of effective salvage therapies. Thus, based on response rates and depth of response as surrogate markers for outcome, 3-drug combinations are, in 2012, the standard of care before ASCT.76 

Bortezomib incorporated into conditioning, consolidation, and maintenance therapy.

As well as being studied in induction regimens, bortezomib has been investigated for use elsewhere within the transplantation treatment paradigm, broadening the utility of PIs in MM. For example, based on the observed synergy with melphalan,48  bortezomib has been examined as part of conditioning regimens. In a phase 2 IFM study of bortezomib plus melphalan 200 mg/m2 (MEL200; the standard of care) as conditioning (Bor-HDM),77  70% of patients achieved at least VGPR, including 32% CR after ASCT. Bortezomib did not increase hematologic toxicity, and only 1 case of grade 3 or 4 PN was reported. A matched control analysis with patients from the IFM2005-01 trial (MEL200 alone) showed the CR rate to be higher in the Bor-HDM group, regardless of induction therapy.77  The results suggest that Bor-HDM is a safe and promising conditioning regimen. These findings were confirmed in a similar United States phase 1 or 2 trial involving 39 patients.78  Nevertheless, randomized studies are needed to assess whether bortezomib-containing conditioning is superior to MEL200 alone.

Currently, novel agents, including bortezomib, are also being tested after ASCT, with the objective of further improving response rate and quality. Single-agent bortezomib consolidation after ASCT has been investigated by the Nordic group,79  in a phase 3 trial in which 370 patients were randomized to receive no treatment or bortezomib. Preliminary results indicated that bortezomib consolidation was feasible; toxicity was low, with 5% grade 3 or 4 PN. The 6-month post-randomization CR/nCR rate was 35% versus 45% with no treatment versus bortezomib (P < .05). This translated into an improvement in median PFS, from 20 to 27 months (P = .04).

Bortezomib has also been combined with IMiDs and dexamethasone after ASCT. In the most striking report, Ladetto et al treated 39 patients who achieved at least VGPR after ASCT with four 28-day cycles of VTD.80  Response was assessed by qualitative nested PCR and quantitative RT-PCR using tumor clone-specific primers. The CR rate increased from 15% after ASCT to 49% after VTD consolidation, and molecular remissions increased from 3% to 18%. With a median follow-up of 65 months after consolidation, only 2 patients in molecular remission had relapsed, and no patient had died from progressive disease.81  This study is the first to document persistent molecular remissions in MM patients receiving ASCT followed by novel agent-based consolidation therapy. Until now, such impressive results had only been reported in the context of myeloablative allogeneic stem cell transplantation. Results from another prospective randomized Italian trial incorporating TD or VTD as induction and as consolidation after tandem ASCT recently confirmed these findings.82  The probability of upgrading from less than CR to CR after consolidation was significantly higher with VTD versus TD, and PFS was also significantly longer. These results support the use of consolidation, including bortezomib-based combinations; however, it should be noted that further response improvements may be seen in some patients after ASCT in the absence of any additional therapy. Thus, additional data from ongoing randomized trials comparing PI/novel agent-based consolidation versus no consolidation have to be awaited before this procedure can be considered a standard of care.

Bortezomib has also been investigated as maintenance post-ASCT in a large phase 3 trial of VAD versus PAD induction, with up to 2 years' maintenance using thalidomide on the VAD arm, or bortezomib on the PAD arm.75  During maintenance with bortezomib, 38% of patients upgraded their post-ASCT response, and a landmark analysis showed a trend for longer duration of response in the maintenance phase using bortezomib versus thalidomide. Another phase 2 study showed upgraded responses with prolonged weekly bortezomib-dexamethasone followed by TD as maintenance therapy after single ASCT,83  with few toxicities. In addition, results of a phase 3 Spanish myeloma group trial suggest a benefit with bortezomib-based maintenance. After induction with VTD, TD, or complex chemotherapy plus bortezomib, patients were randomized after ASCT to 3 years' maintenance with VT, thalidomide, or IFN-α.84  With a median follow-up of 24 months, 2-year PFS was significantly greater with VT maintenance. Grade 1 to 3 PN was observed in 12% and 10% of patients receiving VT and thalidomide maintenance, respectively. Overall, the results indicate that bortezomib-based maintenance may upgrade responses and prolong PFS, but data from a prospective study comparing PI-based maintenance versus no maintenance are currently lacking.

Our understanding of the ubiquitin-proteasome pathway has increased considerably and, with it, the recognition of the important role that proteasome inhibition plays as a highly effective therapeutic strategy in MM. This has stimulated the development of new PIs with distinct efficacy and safety profiles.8  These second-generation PIs also provide potential new options for patients whose disease has become resistant to bortezomib.

Carfilzomib

Of the second-generation PIs, carfilzomib has progressed furthest in clinical development. Clinical studies have demonstrated durable antitumor activity in patients with relapsed/refractory MM9  and, notably, limited neurotoxicity (Table 4). In a large multicenter phase 2 study (PX-171-004) in patients who had received 1 to 3 prior regimens,85  2 carfilzomib dosing regimens were investigated in bortezomib-naive patients and those previously treated with bortezomib. Patients received either carfilzomib 20 mg/m2 for twelve 28-day cycles, or 20 mg/m2 in cycle 1 and 27 mg/m2 in cycles 2 to 12. In 129 bortezomib-naive patients, the ORR was 48% and was better in patients who received the 20/27- versus 20-mg/m2 regimen.85  In the 20/27-mg/m2 group, responses included 2% CR and 27% VGPR. In the 20-mg/m2 group, median TTP was 8.3 months. The most common AEs were fatigue and hematologic toxicity. The risk of PN was low with both regimens despite the fact that approximately 50% of patients had baseline neuropathy. In patients previously treated with bortezomib,86  carfilzomib 20 mg/m2 yielded 1 CR, 1 VGPR, and 4 PRs. Although the response rate was fairly low, median duration of response was 9 months and median TTP was 5.3 months.

An integrated safety analysis of 526 patients with relapsed/refractory MM who were treated in 3 phase 2 studies of carfilzomib 20/27 mg/m2 was recently reported87  and showed that the most common grade more than or equal to 3 AEs were thrombocytopenia (23%), anemia (22%), lymphopenia (18%), pneumonia (11%), and neutropenia (10%). PN was reported infrequently (14% overall) and was generally mild to moderate in severity. Although 72% of patients had grade more than or equal to 2 PN at study entry, only 13% reported treatment-emergent symptoms. Thus, the safety profile of carfilzomib is quite different from that of bortezomib, which is associated with a high risk of PN, albeit that SC administration of bortezomib is associated with a significantly lower risk of PN.42  Preliminary results of another large multicenter phase 2 study of carfilzomib 20 mg/m2 in relapsed/refractory MM (PX-171-003-A1) have recently been reported.88,89  The ORR was 24%, and patients with unfavorable cytogenetics had a 28% ORR compared with 24% in patients with normal or favorable cytogenetics.

Carfilzomib has also been investigated in combination with lenalidomide and low-dose dexamethasone in patients with relapsed/refractory MM.90  A phase 1b study combined carfilzomib 15 to 27 mg/m2 with lenalidomide 10 to 25 mg plus weekly dexamethasone, yielding an ORR of 55%; the most common grade more than or equal to 3 toxicities were hematologic (thrombocytopenia, anemia, and neutropenia). Furthermore, 2 large, randomized, phase 3 trials are ongoing in patients with relapsed/refractory MM. The ASPIRE trial (N = 700) is comparing carfilzomib plus lenalidomide-dexamethasone with lenalidomide-dexamethasone alone in the relapsed setting (primary endpoint: PFS). The FOCUS trial (N = 302) is comparing carfilzomib monotherapy with best supportive care in the relapsed/refractory setting (primary end point: OS). In addition, early-phase studies are investigating carfilzomib in frontline MM in combination with either thalidomide (CARTHADEX)91  or lenalidomide (CRd)92  and dexamethasone (Table 4).

Marizomib

Efficacy and safety data for marizomib are available from phase 1 trials. Results from 2 parallel, phase 1, dose-escalation studies conducted in Australia and the United States in patients with relapsed/refractory MM were recently reported together (Table 5).93  Among 34 patients, 88% had been previously treated with bortezomib, and 71% were bortezomib-refractory. The MTD of marizomib was 0.4 mg/m2 over a 60-minute infusion or 0.5 mg/m2 over a 120-minute infusion. Dose-limiting toxicities included transient hallucinations, cognitive changes, and loss of balance, which were reversible. The most common drug-related AEs included fatigue, gastrointestinal AEs, dizziness, and headache. There was no evidence of PN or thrombocytopenia. Of 15 patients treated in the active dose range (0.4-0.6 mg/m2), 3 demonstrated a PR, all of whom were bortezomib-refractory. These early data suggest that marizomib has a safety profile that is not overlapping with that of other PIs and is active in bortezomib-refractory patients. A twice-weekly regimen of marizomib 0.5 mg/m2 in combination with low-dose dexamethasone is being investigated further.

MLN9708

To date, phase 1 studies of MLN9708 have investigated the safety, tolerability, and preliminary antimyeloma activity of both oral and IV dosing in patients with relapsed/refractory MM and other malignancies (Table 5).94-97  Preliminary data indicate that MLN9708 has promising activity and produces durable responses in heavily pretreated patients. A phase 1 dose-escalation study of twice-weekly oral MLN9708 determined the MTD to be 2.0 mg/m2.97  The study enrolled 56 patients, who had received a median of 4 prior therapies; all patients had received an IMiD, nearly all had prior bortezomib, and approximately 20% had either prior carfilzomib or marizomib. The most common grade more than or equal to 3 AEs were thrombocytopenia (34%), neutropenia (14%), fatigue (9%), and rash (9%). Only 11% of patients developed PN, which was grade 1 or 2. Among response-evaluable patients, the ORR was 13% (1 CR, 5 PR), and responses were durable for up to 16 months. A phase 1 dose-escalation study of weekly oral dosing has also been reported,96  including 28 patients with relapsed/refractory MM who had received a median of 5 prior regimens. No dose-limiting toxicity had been reported at doses up to 3.95 mg/m2; thus, the MTD had not been reached. Similar to twice-weekly dosing, the most common AEs were fatigue and thrombocytopenia. Among 16 response-evaluable patients, 1 patient had a PR and 5 patients had stable disease for up to 10 months. These data suggest that weekly administration of this novel oral PI is well tolerated and has antitumor activity in heavily pretreated relapsed/refractory MM. In addition, as with bortezomib and carfilzomib, MLN9708 is being investigated in a phase 1 or 2 study in frontline MM in combination with lenalidomide and dexamethasone (Table 5).95 

In conclusion, over the past decade, we have moved from the era of proteasome inhibition as a novel therapeutic approach to one in which bortezomib has contributed greatly to improvements in the prognosis of MM. In the past 10 years, clinicians have substantially increased their knowledge of the effects of proteasome inhibition and learned how to use bortezomib highly effectively, including appropriate monitoring for known toxicities, extending its use across multiple areas of the treatment paradigm and successfully introducing a new administrative route. Initially used as single agent in the relapsed setting, this drug has become the backbone of frontline therapy in young and elderly patients, and ongoing studies are exploring new indications, such as consolidation and maintenance. In addition, increasing clinical experience in the use of bortezomib has resulted in the introduction of a new dosing regimen, the weekly schedule, which has demonstrated reduced toxicity while maintaining efficacy. Furthermore, the SC route of administration is now available, which is less neurotoxic, with efficacy comparable with that seen with IV bortezomib. Several second-generation PIs are now in clinical development; and thanks to the understanding we have gained through the development of bortezomib, it is probable that we will be able to move forward more quickly with developing combination regimens incorporating these novel PIs. Results of ongoing trials will prove important in determining their place in therapy.

The authors thank Steve Hill of FireKite for editorial assistance in the development of this manuscript.

This work was supported by the University Hospital Hotel-Dieu, Nantes, France.

Contribution: P.M. wrote the manuscript; and P.G.R., M.C., R.Z.O., J.F.S.M., A.P., and J.-L.H. corrected the paper and approved the final manuscript.

Conflict-of-interest disclosure: P.M., M.C., J.-F.S.M., and A.P. are on the advisory board of Millenium, Janssen, and Onyx; P.G.R. is on the advisory board of Millenium and Nereus; and R.Z.O. is on the advisory board of Millenium and Onyx. J.L.H. declares no competing financial interests.

Correspondence: Philippe Moreau, Hematology Department, University Hospital Hotel-Dieu, 44093 Nantes cedex 01, France; e-mail: philippe.moreau@chu-nantes.fr.

1
Adams
 
J
The proteasome: structure, function, and role in the cell.
Cancer Treat Rev
2003
, vol. 
29
 
Suppl 1
(pg. 
3
-
9
)
2
Ciechanover
 
A
The ubiquitin-proteasome proteolytic pathway.
Cell
1994
, vol. 
79
 
1
(pg. 
13
-
21
)
3
Ciechanover
 
A
The ubiquitin-proteasome pathway: on protein death and cell life.
EMBO J
1998
, vol. 
17
 
24
(pg. 
7151
-
7160
)
4
Groll
 
M
Ditzel
 
L
Lowe
 
J
, et al. 
Structure of 20S proteasome from yeast at 2.4 A resolution.
Nature
1997
, vol. 
386
 
6624
(pg. 
463
-
471
)
5
Obeng
 
EA
Carlson
 
LM
Gutman
 
DM
, et al. 
Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells.
Blood
2006
, vol. 
107
 
12
(pg. 
4907
-
4916
)
6
Dou
 
QP
Li
 
B
Proteasome inhibitors as potential novel anticancer agents.
Drug Resist Updat
1999
, vol. 
2
 
4
(pg. 
215
-
223
)
7
Adams
 
J
The proteasome: a suitable antineoplastic target.
Nat Rev Cancer
2004
, vol. 
4
 
5
(pg. 
349
-
360
)
8
Dick
 
LR
Fleming
 
PE
Building on bortezomib: second-generation proteasome inhibitors as anti-cancer therapy.
Drug Discov Today
2010
, vol. 
15
 
5
(pg. 
243
-
249
)
9
Jain
 
S
Diefenbach
 
C
Zain
 
J
O'Connor
 
OA
Emerging role of carfilzomib in treatment of relapsed and refractory lymphoid neoplasms and multiple myeloma.
Core Evid
2011
, vol. 
6
 (pg. 
43
-
57
)
10
Kupperman
 
E
Lee
 
EC
Cao
 
Y
, et al. 
Evaluation of the proteasome inhibitor MLN9708 in preclinical models of human cancer.
Cancer Res
2010
, vol. 
70
 
5
(pg. 
1970
-
1980
)
11
Boccadoro
 
M
Morgan
 
G
Cavenagh
 
J
Preclinical evaluation of the proteasome inhibitor bortezomib in cancer therapy.
Cancer Cell Int
2005
, vol. 
5
 
1
pg. 
18
 
12
Reddy
 
N
Czuczman
 
MS
Enhancing activity and overcoming chemoresistance in hematologic malignancies with bortezomib: preclinical mechanistic studies.
Ann Oncol
2010
, vol. 
21
 
9
(pg. 
1756
-
1764
)
13
Hideshima
 
T
Richardson
 
PG
Anderson
 
KC
Mechanism of action of proteasome inhibitors and deacetylase inhibitors and the biological basis of synergy in multiple myeloma.
Mol Cancer Ther
2011
, vol. 
10
 
11
(pg. 
2034
-
2042
)
14
Wright
 
JJ
Combination therapy of bortezomib with novel targeted agents: an emerging treatment strategy.
Clin Cancer Res
2010
, vol. 
16
 
16
(pg. 
4094
-
4104
)
15
Zangari
 
M
Terpos
 
E
Zhan
 
F
Tricot
 
G
Impact of bortezomib on bone health in myeloma: a review of current evidence [published online ahead of print January 6, 2012].
Cancer Treat Rev
 
16
Piva
 
R
Ruggeri
 
B
Williams
 
M
, et al. 
CEP-18770: a novel, orally active proteasome inhibitor with a tumor-selective pharmacologic profile competitive with bortezomib.
Blood
2008
, vol. 
111
 
5
(pg. 
2765
-
2775
)
17
Demo
 
SD
Kirk
 
CJ
Aujay
 
MA
, et al. 
Antitumor activity of PR-171, a novel irreversible inhibitor of the proteasome.
Cancer Res
2007
, vol. 
67
 
13
(pg. 
6383
-
6391
)
18
Arastu-Kapur
 
S
Anderl
 
JL
Kraus
 
M
, et al. 
Nonproteasomal targets of the proteasome inhibitors bortezomib and carfilzomib: a link to clinical adverse events.
Clin Cancer Res
2011
, vol. 
17
 
9
(pg. 
2734
-
2743
)
19
Kuhn
 
DJ
Chen
 
Q
Voorhees
 
PM
, et al. 
Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma.
Blood
2007
, vol. 
110
 
9
(pg. 
3281
-
3290
)
20
Chauhan
 
D
Singh
 
AV
Aujay
 
M
, et al. 
A novel orally active proteasome inhibitor ONX 0912 triggers in vitro and in vivo cytotoxicity in multiple myeloma.
Blood
2010
, vol. 
116
 
23
(pg. 
4906
-
4915
)
21
Hurchla
 
MA
Garcia-Gomez
 
A
Hornick
 
MC
, et al. 
Epoxyketone-based proteasome inhibitors carfilzomib and orally bioavailable ONX 0912 have anti-resorptive and bone-anabolic activity in addition to anti-myeloma effects [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
1253
  
Abstract 2906
22
Chauhan
 
D
Catley
 
L
Li
 
G
, et al. 
A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from bortezomib.
Cancer Cell
2005
, vol. 
8
 
5
(pg. 
407
-
419
)
23
Potts
 
C
Albitar
 
X
Anderson
 
C
, et al. 
Marizomib, a proteasome inhibitor for all seasons: preclinical profile and a framework for clinical trials.
Curr Cancer Drug Targets
2011
, vol. 
11
 
3
(pg. 
254
-
284
)
24
Aghajanian
 
C
Soignet
 
S
Dizon
 
DS
, et al. 
A phase I trial of the novel proteasome inhibitor PS341 in advanced solid tumor malignancies.
Clin Cancer Res
2002
, vol. 
8
 
8
(pg. 
2505
-
2511
)
25
Orlowski
 
RZ
Stinchcombe
 
TE
Mitchell
 
BS
, et al. 
Phase I trial of the proteasome inhibitor PS-341 in patients with refractory hematologic malignancies.
J Clin Oncol
2002
, vol. 
20
 
22
(pg. 
4420
-
4427
)
26
Papandreou
 
CN
Daliani
 
DD
Nix
 
D
, et al. 
Phase I trial of the proteasome inhibitor bortezomib in patients with advanced solid tumors with observations in androgen-independent prostate cancer.
J Clin Oncol
2004
, vol. 
22
 
11
(pg. 
2108
-
2121
)
27
Richardson
 
PG
Barlogie
 
B
Berenson
 
J
, et al. 
A phase 2 study of bortezomib in relapsed, refractory myeloma.
N Engl J Med
2003
, vol. 
348
 
26
(pg. 
2609
-
2617
)
28
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
, vol. 
127
 
2
(pg. 
165
-
172
)
29
Jagannath
 
S
Barlogie
 
B
Berenson
 
JR
, et al. 
Updated survival analyses after prolonged follow-up of the phase 2, multicenter CREST study of bortezomib in relapsed or refractory multiple myeloma.
Br J Haematol
2008
, vol. 
143
 
4
(pg. 
537
-
540
)
30
Richardson
 
PG
Sonneveld
 
P
Schuster
 
MW
, et al. 
Bortezomib or high-dose dexamethasone for relapsed multiple myeloma.
N Engl J Med
2005
, vol. 
352
 
24
(pg. 
2487
-
2498
)
31
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.
Blood
2007
, vol. 
110
 
10
(pg. 
3557
-
3560
)
32
Cavaletti
 
G
Gilardini
 
A
Canta
 
A
, et al. 
Bortezomib-induced peripheral neurotoxicity: a neurophysiological and pathological study in the rat.
Exp Neurol
2007
, vol. 
204
 
1
(pg. 
317
-
325
)
33
Csizmadia
 
V
Raczynski
 
A
Csizmadia
 
E
, et al. 
Effect of an experimental proteasome inhibitor on the cytoskeleton, cytosolic protein turnover, and induction in the neuronal cells in vitro.
Neurotoxicology
2008
, vol. 
29
 
2
(pg. 
232
-
243
)
34
Silverman
 
L
Csizmadia
 
V
Brewer
 
K
Simpson
 
C
Alden
 
C
Proteasome inhibitor associated neuropathy is mechanism based [abstract].
Blood (ASH Annual Meeting Abstracts)
2008
, vol. 
112
 
11
pg. 
2646a
  
Abstract 2646
35
Delforge
 
M
Blade
 
J
Dimopoulos
 
MA
, et al. 
Treatment-related peripheral neuropathy in multiple myeloma: the challenge continues.
Lancet Oncol
2010
, vol. 
11
 
11
(pg. 
1086
-
1095
)
36
Mohty
 
B
El-Cheikh
 
J
Yakoub-Agha
 
I
, et al. 
Peripheral neuropathy and new treatments for multiple myeloma: background and practical recommendations.
Haematologica
2010
, vol. 
95
 
2
(pg. 
311
-
319
)
37
Richardson
 
PG
Delforge
 
M
Beksac
 
M
, et al. 
Management of treatment-emergent peripheral neuropathy in multiple myeloma.
Leukemia
2012
, vol. 
26
 
4
(pg. 
595
-
608
)
38
Sonneveld
 
P
Jongen
 
JL
Dealing with neuropathy in plasma-cell dyscrasias.
Hematology Am Soc Hematol Educ Program
2010
(pg. 
423
-
430
)
39
Bringhen
 
S
Larocca
 
A
Rossi
 
D
, et al. 
Efficacy and safety of once-weekly bortezomib in multiple myeloma patients.
Blood
2010
, vol. 
116
 
23
(pg. 
4745
-
4753
)
40
Mateos
 
MV
Oriol
 
A
Martinez-Lopez
 
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
, vol. 
11
 
10
(pg. 
934
-
941
)
41
Palumbo
 
A
Bringhen
 
S
Rossi
 
D
, et al. 
Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: a randomized controlled trial.
J Clin Oncol
2010
, vol. 
28
 
34
(pg. 
5101
-
5109
)
42
Moreau
 
P
Pylypenko
 
H
Grosicki
 
S
, et al. 
Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study.
Lancet Oncol
2011
, vol. 
12
 
5
(pg. 
431
-
440
)
43
San Miguel
 
J
Blade
 
J
Boccadoro
 
M
, et al. 
A practical update on the use of bortezomib in the management of multiple myeloma.
Oncologist
2006
, vol. 
11
 
1
(pg. 
51
-
61
)
44
Lonial
 
S
Waller
 
EK
Richardson
 
PG
, et al. 
Risk factors and kinetics of thrombocytopenia associated with bortezomib for relapsed, refractory multiple myeloma.
Blood
2005
, vol. 
106
 
12
(pg. 
3777
-
3784
)
45
Moreau
 
P
Coiteux
 
V
Hulin
 
C
, et al. 
Prospective comparison of subcutaneous versus intravenous administration of bortezomib in patients with multiple myeloma.
Haematologica
2008
, vol. 
93
 
12
(pg. 
1908
-
1911
)
46
Hrusovsky
 
I
Emmerich
 
B
von Rohr
 
A
, et al. 
Bortezomib retreatment in relapsed multiple myeloma: results from a retrospective multicentre survey in Germany and Switzerland.
Oncology
2010
, vol. 
79
 
3-4
(pg. 
247
-
254
)
47
Petrucci
 
MT
Blau
 
I
Corradini
 
P
, et al. 
Efficacy and safety of retreatment with bortezomib in patients with multiple myeloma: interim results from RETRIEVE, a prospective international phase 2 study [abstract].
Haematologica
2010
, vol. 
95
 
s2
152
 
Abstract 0377
48
Mitsiades
 
N
Mitsiades
 
CS
Richardson
 
PG
, et al. 
The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications.
Blood
2003
, vol. 
101
 
6
(pg. 
2377
-
2380
)
49
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
, vol. 
105
 
8
(pg. 
3058
-
3065
)
50
Orlowski
 
RZ
Nagler
 
A
Sonneveld
 
P
, et al. 
Randomized phase III study of pegylated liposomal doxorubicin plus bortezomib compared with bortezomib alone in relapsed or refractory multiple myeloma: combination therapy improves time to progression.
J Clin Oncol
2007
, vol. 
25
 
25
(pg. 
3892
-
3901
)
51
Horton
 
TM
Gannavarapu
 
A
Blaney
 
SM
, et al. 
Bortezomib interactions with chemotherapy agents in acute leukemia in vitro.
Cancer Chemother Pharmacol
2006
, vol. 
58
 
1
(pg. 
13
-
23
)
52
Jagannath
 
S
Richardson
 
PG
Barlogie
 
B
, et al. 
Bortezomib in combination with dexamethasone for the treatment of patients with relapsed and/or refractory multiple myeloma with less than optimal response to bortezomib alone.
Haematologica
2006
, vol. 
91
 
7
(pg. 
929
-
934
)
53
Mikhael
 
JR
Belch
 
AR
Prince
 
HM
, et al. 
High response rate to bortezomib with or without dexamethasone in patients with relapsed or refractory multiple myeloma: results of a global phase 3b expanded access program.
Br J Haematol
2009
, vol. 
144
 
2
(pg. 
169
-
175
)
54
Garderet
 
L
Iacobelli
 
S
Moreau
 
P
, et al. 
Bortezomib (VELCADE)-thalidomide-dexamethasone (VTD) is superior to thalidomide-dexamethasone (TD) in patients with multiple myeloma (MM) progressing or relapsing after autologous transplantation [abstract].
Haematologica
2011
, vol. 
96
 
s2
(pg. 
420
-
421
Abstract 1008
55
Dimopoulos
 
MA
Jagannath
 
S
Yoon
 
S-S
, et al. 
Vantage 088. Vorinostat in combination with bortezomib in patients with relapsed/refractory multiple myeloma: results of a global, randomized phase 3 trial [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
(pg. 
368
-
369
Abstract 811
56
Lonial
 
S
Mitsiades
 
CS
Richardson
 
PG
Treatment options for relapsed and refractory multiple myeloma.
Clin Cancer Res
2011
, vol. 
17
 
6
(pg. 
1264
-
1277
)
57
Mateos
 
MV
Hernandez
 
JM
Hernandez
 
MT
, et al. 
Bortezomib plus melphalan and prednisone in elderly untreated patients with multiple myeloma: results of a multicenter phase 1/2 study.
Blood
2006
, vol. 
108
 
7
(pg. 
2165
-
2172
)
58
Mateos
 
MV
Hernandez
 
JM
Hernandez
 
MT
, et al. 
Bortezomib plus melphalan and prednisone in elderly untreated patients with multiple myeloma: updated time-to-events results and prognostic factors for time to progression.
Haematologica
2008
, vol. 
93
 
4
(pg. 
560
-
565
)
59
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
, vol. 
359
 
9
(pg. 
906
-
917
)
60
Mateos
 
MV
Richardson
 
PG
Schlag
 
R
, et al. 
Bortezomib plus melphalan and prednisone compared with melphalan and prednisone in previously untreated multiple myeloma: updated follow-up and impact of subsequent therapy in the phase III VISTA trial.
J Clin Oncol
2010
, vol. 
28
 
13
(pg. 
2259
-
2266
)
61
San Miguel
 
JF
Schlag
 
R
Khuageva
 
NK
, et al. 
Continued overall survival benefit after 5 years' follow-up with bortezomib-melphalan-prednisone (VMP) versus melphalan-prednisone (MP) in patients with previously untreated multiple myeloma, and no increased risk of second primary malignancies: final results of the phase 3 VISTA trial [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
(pg. 
221
-
222
Abstract 476
62
Mateos
 
MV
Oriol
 
A
Teruel
 
AI
, et al. 
Maintenance therapy with bortezomib plus thalidomide (VT) or bortezomib plus prednisone (VP) in elderly myeloma patients included in the GEM2005MAS65 Spanish randomized trial [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
222
  
Abstract 477
63
Corso
 
A
Barbarano
 
L
Mangiacavalli
 
S
, et al. 
Bortezomib plus dexamethasone can improve stem cell collection and overcome the need for additional chemotherapy before autologous transplant in patients with myeloma.
Leuk Lymphoma
2010
, vol. 
51
 
2
(pg. 
236
-
242
)
64
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
, vol. 
91
 
11
(pg. 
1498
-
1505
)
65
Jagannath
 
S
Durie
 
BG
Wolf
 
JL
, et al. 
Extended follow-up of a phase 2 trial of bortezomib alone and in combination with dexamethasone for the frontline treatment of multiple myeloma.
Br J Haematol
2009
, vol. 
146
 
6
(pg. 
619
-
626
)
66
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.
J Clin Oncol
2010
, vol. 
28
 
30
(pg. 
4621
-
4629
)
67
Wang
 
M
Giralt
 
S
Delasalle
 
K
Handy
 
B
Alexanian
 
R
Bortezomib in combination with thalidomide-dexamethasone for previously untreated multiple myeloma.
Hematology
2007
, vol. 
12
 
3
(pg. 
235
-
239
)
68
Jakubowiak
 
AJ
Kendall
 
T
Al-Zoubi
 
A
, et al. 
Phase II trial of combination therapy with bortezomib, pegylated liposomal doxorubicin, and dexamethasone in patients with newly diagnosed myeloma.
J Clin Oncol
2009
, vol. 
27
 
30
(pg. 
5015
-
5022
)
69
Popat
 
R
Oakervee
 
HE
Hallam
 
S
, et al. 
Bortezomib, doxorubicin and dexamethasone (PAD) front-line treatment of multiple myeloma: updated results after long-term follow-up.
Br J Haematol
2008
, vol. 
141
 
4
(pg. 
512
-
516
)
70
Richardson
 
PG
Weller
 
E
Lonial
 
S
, et al. 
Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma.
Blood
2010
, vol. 
116
 
5
(pg. 
679
-
686
)
71
Reeder
 
CB
Reece
 
DE
Kukreti
 
V
, et al. 
Once- versus twice-weekly bortezomib induction therapy with CyBorD in newly diagnosed multiple myeloma.
Blood
2010
, vol. 
115
 
16
(pg. 
3416
-
3417
)
72
Cavo
 
M
Tacchetti
 
P
Patriarca
 
F
, et al. 
Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study.
Lancet
2010
, vol. 
376
 
9758
(pg. 
2075
-
2085
)
73
Moreau
 
P
Avet-Loiseau
 
H
Facon
 
T
, et al. 
Bortezomib plus dexamethasone versus reduced-dose bortezomib, thalidomide plus dexamethasone as induction treatment before autologous stem cell transplantation in newly diagnosed multiple myeloma.
Blood
2011
, vol. 
118
 
22
(pg. 
5752
-
5758
)
74
Rosiñol
 
L
Cibeira
 
MT
Mateos
 
MV
, et al. 
Results of pre- and post-autologous stem cell transplantation (ASCT) with three induction regimens in multiple myeloma (MM): superiority of VTD (bortezomib/thalidomide/dexamethasone) over TD and VBMCP/VBAD plus bortezomib (VBMCP/VBAD/V) [abstract].
Haematologica
2011
, vol. 
96
 
s1
S69
 
Abstract P-138
75
Sonneveld
 
P
Schmidt-Wolf
 
I
van der Holt
 
B
, et al. 
HOVON-65/GMMG-HD4 randomized phase III trial comparing bortezomib, doxorubicin, dexamethasone (PAD) vs VAD followed by high-dose melphalan (HDM) and maintenance with bortezomib or thalidomide in patients with newly diagnosed multiple myeloma (MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2010
, vol. 
116
 
21
(pg. 
23
-
24
Abstract 40
76
Cavo
 
M
Rajkumar
 
SV
Palumbo
 
A
, et al. 
International Myeloma Working Group consensus approach to the treatment of multiple myeloma patients who are candidates for autologous stem cell transplantation.
Blood
2011
, vol. 
117
 
23
(pg. 
6063
-
6073
)
77
Roussel
 
M
Moreau
 
P
Huynh
 
A
, et al. 
Bortezomib and high-dose melphalan as conditioning regimen before autologous stem cell transplantation in patients with de novo multiple myeloma: a phase 2 study of the Intergroupe Francophone du Myelome (IFM).
Blood
2010
, vol. 
115
 
1
(pg. 
32
-
37
)
78
Lonial
 
S
Kaufman
 
J
Tighiouart
 
M
, et al. 
A phase I/II trial combining high-dose melphalan and autologous transplant with bortezomib for multiple myeloma: a dose- and schedule-finding study.
Clin Cancer Res
2010
, vol. 
16
 
20
(pg. 
5079
-
5086
)
79
Mellqvist
 
UH
Gimsing
 
P
Hjertner
 
O
, et al. 
Improved progression-free survival with bortezomib consolidation after high dose melphalan: results of a randomized phase III trial [abstract].
Haematologica
2011
, vol. 
96
 
s1
S31
 
Abstract O-11
80
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.
J Clin Oncol
2010
, vol. 
28
 
12
(pg. 
2077
-
2084
)
81
Ladetto
 
M
Ferrero
 
S
Drandi
 
D
, et al. 
Long-term results of the GIMEMA VTD consolidation trial in autografted multiple myeloma patients (VEL-03-096): impact of minimal residual disease detection by real time quantitative PCR on late recurrences and overall survival [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
(pg. 
376
-
377
Abstract 827
82
Cavo
 
M
Pantani
 
L
Patriarca
 
F
, et al. 
Superior complete response rate (CR) and progression-free survival (PFS) with bortezomib-thalidomide-dexamethasone (VTD) versus thalidomide-dexamethasone (TD) as consolidation therapy after autologous stem-cell transplantation (ASCT) in multiple myeloma (MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
816
  
Abstract 1871
83
Sahebi
 
F
Frankel
 
PH
Farol
 
L
, et al. 
Sequential bortezomib, dexamethasone, and thalidomide maintenance therapy after single autologous peripheral stem cell transplantation in patients with multiple myeloma.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
3
(pg. 
486
-
492
)
84
Rosiñol
 
L
Cibeira
 
MT
Mateos
 
MV
, et al. 
A phase III PETHEMA/GEM randomized trial of postransplant (ASCT) maintenance in multiple myeloma: superiority of bortezomib/thalidomide compared with thalidomide and alfa-2b interferon [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
(pg. 
1694
-
1695
Abstract 3962
85
Vij
 
R
Wang
 
M
Kaufman
 
JL
, et al. 
An open-label, single-arm, phase 2 (PX-171-004) study of single-agent carfilzomib in bortezomib-naive patients with relapsed and/or refractory multiple myeloma [published online ahead of print May 3, 2012].
Blood
 
86
Vij
 
R
Siegel
 
DS
Kaufman
 
JL
, et al. 
Results of an ongoing open-label, phase II study of carfilzomib in patients with relapsed and/or refractory multiple myeloma (R/R MM) [abstract].
J Clin Oncol
2010
, vol. 
28
 
15s
pg. 
573s
  
Abstract 8000
87
Singhal
 
S
Siegel
 
DS
Martin
 
T
, et al. 
Integrated safety from phase 2 studies of monotherapy carfilzomib in patients with relapsed and refractory multiple myeloma (MM): an updated analysis [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
819
  
Abstract 1876
88
Siegel
 
DS
Martin
 
T
Wang
 
M
, et al. 
Results of PX-171-003-A1, an open-label, single-arm, phase 2 (ph 2) study of carfilzomib (CFZ) in patients (pts) with relapsed and refractory multiple myeloma (MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2010
, vol. 
116
 
21
pg. 
433
  
Abstract 985
89
Jakubowiak
 
AJ
Martin
 
T
Singhal
 
S
, et al. 
Responses to single-agent carfilzomib (CFZ) are not affected by cytogenetics in patients (pts) with relapsed and refractory multiple myeloma (R/R MM) [abstract].
Ann Oncol
2011
, vol. 
22
 
Suppl 4
pg. 
iv122
  
Abstract 117
90
Niesvizky
 
R
Wang
 
L
Orlowski
 
RZ
, et al. 
Phase Ib multicenter dose escalation study of carfilzomib plus lenalidomide and low dose dexamethasone (CRd) in relapsed and refractory multiple myeloma (MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
(pg. 
128
-
129
Abstract 304
91
Sonneveld
 
P
Hacker
 
E
Zweegman
 
S
, et al. 
Carfilzomib combined with thalidomide and dexamethasone (CARTHADEX) as induction treatment prior to high-dose melphalan (HDM) in newly diagnosed patients with multiple myeloma (MM): a trial of the European Myeloma Network EMN [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
290
  
Abstract 633
92
Jakubowiak
 
AJ
Dytfeld
 
D
Jagannath
 
S
, et al. 
Final results of a frontline phase 1/2 study of carfilzomib, lenalidomide, and low-dose dexamethasone (CRd) in multiple myeloma (MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
12
(pg. 
288
-
289
Abstract 631
93
Richardson
 
PG
Spencer
 
A
Cannell
 
P
, et al. 
Phase 1 clinical evaluation of twice-weekly marizomib (NPI-0052), a novel proteasome inhibitor, in patients with relapsed/refractory multiple myeloma (MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
(pg. 
140
-
141
Abstract 302
94
Assouline
 
S
Chang
 
J
Rifkin
 
R
, et al. 
MLN9708, a novel, investigational proteasome inhibitor, in patients with relapsed/refractory lymphoma: results of a phase 1 dose-escalation study [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
1148
  
Abstract 2672
95
Berdeja
 
JG
Richardson
 
PG
Lonial
 
S
, et al. 
Phase 1/2 study of oral MLN9708, a novel, investigational proteasome inhibitor, in combination with lenalidomide and dexamethasone in patients with previously untreated multiple myeloma (MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
223
  
Abstract 479
96
Kumar
 
S
Bensinger
 
WI
Reeder
 
CB
, et al. 
Weekly dosing of the investigational oral proteasome inhibitor MLN9708 in patients with relapsed and/or refractory multiple myeloma: results from a phase 1 dose-escalation study [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
(pg. 
371
-
372
Abstract 816
97
Richardson
 
PG
Baz
 
R
Wang
 
L
, et al. 
Investigational agent MLN9708, an oral proteasome inhibitor, in patients (pts) with relapsed and/or refractory multiple myeloma (MM): results from the expansion cohorts of a phase 1 dose-escalation study [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
140
  
Abstract 301
98
Richardson
 
PG
Barlogie
 
B
Berenson
 
J
, et al. 
Extended follow-up of a phase II trial in relapsed, refractory multiple myeloma: final time-to-event results from the SUMMIT trial.
Cancer
2006
, vol. 
106
 
6
(pg. 
1316
-
1319
)
99
Niesvizky
 
R
Flinn
 
IW
Rifkin
 
R
, et al. 
Efficacy and safety of three bortezomib-based combinations in elderly, newly diagnosed multiple myeloma patients: results from all randomized patients in the community-based, phase 3b UPFRONT study [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
(pg. 
222
-
223
Abstract 478
100
Jagannath
 
S
Vij
 
R
Stewart
 
AK
, et al. 
Final results of PX-171-003-A0, part 1 of an open-label, single-arm, phase II study of carfilzomib (CFZ) in patients (pts) with relapsed and refractory multiple myeloma (MM) [abstract].
J Clin Oncol
2009
, vol. 
27
 
15s
pg. 
435s
  
Abstract 8504
Sign in via your Institution