Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jul 1.
Published in final edited form as: Semin Hematol. 2012 Jul;49(3):228–242. doi: 10.1053/j.seminhematol.2012.04.010

Bortezomib Combination Therapy in Multiple Myeloma

Prashant Kapoor 1, Vijay Ramakrishnan 1, S Vincent Rajkumar 1
PMCID: PMC3597231  NIHMSID: NIHMS380927  PMID: 22726546

Abstract

Bortezomib was approved for the treatment of multiple myeloma in 2003. Since then several bortezomib-based combination therapies have emerged. Although some combinations have been preceded by preclinical investigations, most have followed the inevitable process in which active (or potentially active) drugs are combined with each other to create new treatment regimens. Regimens that have combined bortezomib with corticosteroids, alkylating agents, thalidomide, and/or lenalidomide have resulted in high response rates. Despite the higher and often deeper response rates and prolongation of progression-free survival with bortezomib-based multiagent regimens, an overall survival (OS) advantage has not been demonstrated with most combinations compared to the sequential approach of utilizing anti-myeloma agents, particularly in patients less than 65 with newly diagnosed myeloma. The unique properties of some of these regimens can be taken into account when choosing a particular regimen based on the clinical scenario. For example, bortezomib, thalidomide, dexamethasone (VTD) has particular value in renal failure since none of the drugs need dose modification. Similarly, the combination chemotherapy regimen VDT-PACE (bortezomib, dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide, etoposide) is of particular value in patients presenting with aggressive disease such as extramedullary plasmacytomas or plasma cell leukemia. Ongoing clinical trials are testing combinations of bortezomib with several other classes of agents, including monoclonal antibodies, and inhibitors of deacetylases, heat shock proteins, phosphatidyl inositol 3-kinase/Akt/mammalian target of rapamycin pathway and farnesyl transferase.

Introduction

Bortezomib is boronate-based dipeptide proteasome inhibitor (PI) that primarily targets the chymotrypsin-like activities of the intracellular proteasome enzyme complex.1 It received accelerated approval by the US Food and Drug Administration in the year 2003 based on a large multicenter phase II clinical trial.2 This trial demonstrated an overall response in nearly a third of patients with advanced multiple myeloma (MM). Subsequent studies have attributed improvement in overall survival (OS) of MM patients in the last decade to the use of bortezomib, as well as other agents such as thalidomide and lenalidomide that are commonly referred to as immunomodulatory drugs (IMiDs). However, most, if not all, patients inevitably relapse. Each relapse requires salvage therapy, and there is decreasing response duration with successive lines of salvage therapies. Although the activity of bortezomib has been demonstrated with retreatment in prior responders, the median OS of patients who become refractory to bortezomib and IMiDs is disappointingly short (~ 9 months).3

Bortezomib initially showed activity in a phase 1 trial in which clinical benefit was noted in all 9 heavily pre-treated MM patients.4 Subsequently, the phase 2 SUMMIT (Study of Uncontrolled Multiple Myeloma Managed with Proteasome Inhibition Therapy) and CREST (Clinical Response and Efficacy Study of Bortezomib in the Treatment of Relapsing MM) trials demonstrated meaningful benefit in relapsed refractory MM (median TTP 7-11 months and OS 17-60 months).5,6. The phase III APEX (Assessment of Proteasome Inhibition for Extending Remissions) trial which led to the full approval of bortezomib in 2005 in patients who have received at least one prior therapy, demonstrated a clear 6-month survival advantage with bortezomib (median OS 29.8 months) compared to dexamethasone despite cross-over from the dexamethasone arm.7 Here, we review bortezomib-based combination strategies, other than bortezomib-steroid doublets that have been effectively utilized for optimization of clinical response and disease control, particularly in relapsed refractory MM patients who have exhausted the standard therapies or those who are unable to derive ASCT-associated survival benefit owing to their transplant ineligibility status.

Rationale for using bortezomib-based combinations

Although some combinations have been preceded by preclinical investigations, most have followed the inevitable process in which active (or potentially active) drugs in a given malignancy are combined with each other to create new treatment regimens. Nevertheless, a review of possible biological mechanisms through which the activity of bortezomib can be influenced and targeted is worth some discussion. Bortezomib is a prototypical PI that reversibly inhibits the ubiquitin proteasome pathway (UPP) leading to cell-cycle arrest and apoptosis.1 Combinations should ideally take into account the different mechanisms of action of bortezomib, drug resistance pathways, and incorporate strategies designed to improve sensitivity of myeloma cells to the drug. The molecular mechanisms of proteasome inhibition and the preclinical activity of bortezomib-based combinations have been elucidated in detail elsewhere in this issue of Seminars.

Compared to normal cells, neoplastic plasma cells are more dependent on a functional unfolded protein response (UPR) for proper folding of abundantly synthesized immunoglobulins. Bortezomib and other PIs inhibit the UPP which results in the accumulation of misfolded proteins leading to UPR-induced cell death.8 In addition bortezomib inhibits plasma-cell adhesion to the microenvironmental bone marrow stromal cells (BMSCs) and angiogenesis.1 The complementary and/or augmenting effects of various agents used in bortezomib-based combinations have been demonstrated in preclinical studies (Table1). The most compelling rationale for many of the combinations is from clinical trials that have exploited the additive or synergistic anti-neoplastic effect that arises when one combines drug classes with impressive independent single-agent activity and unique non-overlapping toxicities. Bortezomib lends itself to multi-drug combinations since its myelosuppressive effect is typically mild, transient and non-cumulative. We have discussed below several bortezomib-based combinations with promising clinical activities.

Table 1.

Rationale for Novel Combinations with Bortezomib (V)

Class Drug(s) Rationale for Combination Therapy with Bortezomib
Enzyme Inhibitors
HSP inhibitors Tanespimycin Resistance to V is mediated via upregulation of HSPs. HSPIs sensitize MM cells to V by inhibiting HSP which then leads to increased accumulation of misfolded proteins and further activation of endoplasmic stress response.
Deacetylase Inhibitors (DACi) Vorinostat Panabinostat Belinostat Romidepsin Synergy with V has been primarily attributed to the dual inhibition of aggresomes and proteasomes by DACi and V, respectively, thereby blocking protein degradation by different mechanisms. Upregulation of CDK inhibitors such as p21 (vorinostat) or down regulation of antiapoptotic Bcl2 (romidepsin) are some specific mechanisms that have been elucidated.
AKT/mTOR inhibitors Perifosine Temsirolimus Treatment with V has been shown to upregulate pAkt in MM cells and perifosine has been shown to inhibit the upregulated pAkt. In addition, perifosine enhances V induced cytotoxicity via activation of JNK (a stress response protein mediating apoptosis by UPR).
Pan-Bcl inhibitors Obatoclax Antagonist of BH3 binding groove of Bcl family proteins; induces apoptosis by interfering with the interaction between prov.(e.g.Bak)and anti-apoptotic proteins (e.g.Mcl-1 which accumulates with V therapy a a result of PI) demonstrating augmented cytotoxic responses- against MM cells
Farnesyl Transferase Inhibitors(FTIs) Tipifarnib Lonafarnib FTIs in combination with V overcome CAM-DR; induce ER stress-related MM cell apoptosis, downregulation of pAKT and disruption of aggresome complex via inhibition of HDAC6
Immunomodulating Agents (IMiDs) Thalidomide Lenalidomide Pomalidomide R, in particular, triggers caspase-8 dependent apoptosis and sensitizes MM cells to death receptor, Fas-mediated apoptosis; disrupts BMSC support, downregulates NFKB by inhibition of IGF-1 and TNF-α (in contrast to IKKB mediated downregulation by V); immunomodulation with improved NK cell- mediated MM cell destruction.
DNA Damaging Agents Alkylators Anthracyclines Bendamustine In addition to its proapoptotic effects through decreased expression of Bcl-2, XIAP and other antiapoptotic factors V inhibits the protective cellular response to genotoxic stress thereby restoring the sensitivity to DNA damaging agents. Anthracyclines further demonstrate synergy by suppress V-induced HSP expression
Monoclonal Antibodies Elotuzumab Mapatumumab Siltuximabb V enhances activity of monoclonal antibodies by rendering MM cells more vulnerable to NK cell -mediated ADCC

Abbreviations: V=Bortezomib, HSPi = Heat Shock Protein Inhibitor, IGs = Immunoglobulins, PI= Proteasome Inhibitor, JNK= C-Jun N terminal kinase, UPR=Unfolded Protein Response, CAM-DR = Cell-Adhesion Mediated Drug Resistance, R=Lenalidomide, IGF-1 Insulin like Growth Factor, TNF-α = Tumor Necrosis Factor Alpha, ADCC Antibody-Dependent Cellular Cytotoxicity

A. Immunomodulatory Drugs (IMiDs)-Bortezomib-based combinations

a. Lenalidomide-Bortezomib

The mechanism of anti-myeloma activity of lenalidomide is different from that of bortezomib although some degree of overlap exists. The value of this combination has been prospectively assessed in a clinical phase I/II study of bortezomib-lenalidomide-dexamethasone (VRD) administered as 3-week cycle in newly diagnosed patients).9 Sensory neuropathy (80%) and fatigue (54%) were commonly seen although the regimen was not associated with any treatment-related deaths. An unprecedented 100% overall response rate (ORR) was seen, with nearly three-fourths of the phase II cohort achieving a very good partial response (VGPR) or better. Despite the impressive initial response rates, a quarter of the patients relapsed within eighteen months.9 This highly effective regimen has also been studied in the relapsed-refractory setting (Table 2: note that the dose is different in relapsed setting) in which 84% patients achieved minimal response (MR) or better with a median duration of response of 24 weeks (range 6-81).10 The relatively small sample size of high-risk patients precludes one from drawing definite conclusions about the efficacy of this combination in patients with adverse cytogenetics. The high response rates with VRD have led to the premature adoption of this regimen as a standard front-line treatment outside of clinical trials in the United States. For standard- and intermediate-risk patients with newly diagnosed myeloma, we do not recommend VRD since there are no phase III data to support such therapy. However, in newly diagnosed patients with high-risk features for whom attainment and maintenance of a complete response (CR) is an important therapeutic goal, VRD is a reasonable alternative to other standard regimens. VRD is also of value in patients with relapsed refractory myeloma.10

Table 2.

Selected Phase 1 /2 Trials incorporating Bortezomib + IMiDs and/or Cytotoxic agents

Study Regimen Cohort N ORR ≥VGPR (%) MTD/Phase 2 dose Cycle
Days #

EVOLUTION VDCR NDMM ±ASCT 25 96 56 V 1.3mg/m2 IV d 1,4,8,11
D 40mg PO d 1,8,15
R 15mg (25mg in VDR) PO d 1-14
C 500mg/m2 PO d 1,8 ± d 15 (mod)
V maint. 1.3mg/m2 IV d 1,8, 15,22
Phase 156 21 8


EVOLUTION VDCR 132 59
Phase 211 VDR 50
VDC 41 42 4
VDC-modified 59

Phase 224,25 CyBorD NDMM ±ASCT 33 88 61 V 1.3mg/m2 IV d 1,4,8,11
D 40mg PO d 1-4,9-12,17-20
C 300mg/m2 PO d 1,8 15,22
V 1.5mg/m2 IV d1,8,15,22 (mod)
D 40mg d 1,8,15,22 from cycle 3(mod)
28 4
CyBorD-modified 30 93 60
Total 63 90 60

 Phase 1/29 RVD NDMM ±ASCT 66 100 67 R 25mg PO d 1-14
V 1.3mg/m2 IV d 1,4,8,11
D 20mg PO d 1,2,4,5,8,9,11,12 cycles 1-4
D 10mg PO d 1,2,4,5,8,9,11,12 cycles 5-8
21 8

Phase 1/257 RVDD NDMM ±ASCT 74 96 67 R 25mg PO d 1-14
V 1.3mg/m2 IV d 1,4,8,11
D 20mg PO d1,2,4,5,8,9,11,12 cycles 1-4
D 10mg PO d 1,2,4,5,8,9,11,12 cycles 5-8 PLD 30mg/m2 IV d4
21 8

DSSM XI VDC NDMM +ASCT 30 77 NA V 1.3mg/m2 IV d 1,4,8,11
D 40mg PO d 1,2,4,5,8,9,11,12
C 900mg/m2 IV d1
21 3
Phase 158 92 @ MTD

Phase 259 VDT (D=PLD) NDMM ±ASCT 40 78 35(CR+nCR) V 1.3mg/m2 IV d 1,4,15,18
PLD 20mg/m2 IV d1,15
T 200mg PO daily continuously
28 6 + 2

Phase 228 DVD NDMM 35 72 29 D 40mg IV d 1,4,8,11
V 1 mg/m2 IV d 1,4,8,11
PLD 5mg/m2 d 1,4,8,11
28 8

Phase 229 VDD NDMM ±ASCT 40 93 63 V 1.3mg/m2 IV d 1,4,8,11
D 40mg PO d1,2,4,5,8,9,11,12 cycles 1
D 20mg PO d 1,2,4,5,8,9,11,12 cycles 2-8
PLD 30mg/m2 IV d4
21 6

Phase 260 DBd
B=bortezomib
NDMM +ASCT 50 98 ≥MR 27(CR+nCR) V 1.3mg/m2 IV d 1,4,8,11
D 40mg PO d1-,4, 8-11, 15-18 cycle 1
D 40mg PO d 1-,4, cycles 2-4
PLD 30mg/m2 IV d4
21 3-4

Phase 1/230 PAD1 NDMM +ASCT 21 95 62 V 1.3mg/m2 IV d 1,4,8,11 (PAD1)
V 1 mg/m2 IV d 1,4,8,11 (PAD1)
D 40mgPO, d1-4,8-11,15-18 cycle1
D 40mgPO, d1-4,cycle2
Dox 9mg/m2 d1-4
21 4
Phase 2 PAD2 20 89 42

Phase 2 VDT-PACE NDMM + tandem ASCT 303 92* NA V 1mg/m2/d IV 1,4,8,11
D 40mg PO, d1-4
T 200mg PO /d continuously
P 10mg/m2/d CIV 1-4
A 10mg/m2/d CIV 1-4
C 400mg/m2/d CIV 1-4
E 40mg/m2/d CIV 1-4
(Interferon± D maintenance in TT2)
2(I)
TT3 VTD (1yr) TD (yr 2 & 3) 668 81(+T) 2(C)
TT234 VAD-DCEP-CAD-DCEP 79(no T) 4 (I)
DPACE/Interferon ±T 4 (C)

Phase 236 VDT-PACE NDMM + tandem ASCT 303 59** NA V 1mg/m2/d IV 1,4,8,11
D 40mg PO, d1-4
T 200mg PO /d continuously
P 10mg/m2/d CIV 1-4
A 10mg/m2/d CIV 1-4
C 400mg/m2/d CIV 1-4
E 40mg/m2/d CIV 1-4
28-42 4
TT3 VTD (1yr) TD (yr 2 & 3)
TT3B VDTPACE VRD (3 yrs) 177 61

Phase 210 VRD RR 64 84 ≥MR 21(CR/nCR) V= 1mg/m2 IVdays 1,4,8,11
V=days 1,8 after cycle 8
R=15mg/day PO days 1-14
D(20-40mg PO days of and afterV
D=10mg 1,2, 8,9 after cycle8
8 21

Phase 1/ 261 VCP RR 37 95 ≥MR 50(CR/nCR) V 1.3mg/m2 IV d 1,4,8,11 or
V 1.5mg/m2 IV d 1,8,15
C 300mg/m2 PO d 1,8 15,22
P 100mg PO every other day
28 8

Phase 262 VDC RR 54 76 15(CR) V 1.3mg/m2 IV d 1,4,8,11 cycle1-8
D 20mg PO d1,2,4,5,8,9,11,12
C 50mg daily PO continuously
V 1.3mg/m2 d1,8,15,22 cycle 9-11)
21 8
35 3

Phase 263 PAd RR 64 67 25 V 1.3mg/m2 IV d 1,4,8,11
D 40mg d1-4
Dox 30mg/m2 IV d1
28 6

Phase 227 VDT RR 28 36 21 V 1 mg/m2 IV d 1,4,8,11
D 24mg PO d1,2,4,5,8,9,11,12
T 100mg PO continuously
28 6
VDT+PLD 42 74 52(CR/nCR)

Phase1/264 V+ Bendamustine RR 39 48 8 V 1 mg/m2 IV d 1,4,8,11
Benda 90mg/m2 IV d 1,4
28 8
*

2-year sustained CR rate I=Induction, C=consolidation

**

2-year CR estimate

Abbreviations: N=Number of patients, ORR=Overall Response Rate, VGPR=Very Good Partial Response, MTD=Maximum tolerated dose V/Bor =Bortezomib, D/d=Dexamethasone, C/Cy=Cyclophosphamide, R=lenalidomide,T=thalidomide, PLD/D=Pegylated Liposomal Doxorubicin, P=Prednisone or CisPlatin (in VDTPACE), E=Etoposide, PAD=Bortezomib, Doxorubicin (Adriamycin), Dexamethasone, NDMM=Newly Diagnosed Multiple Myeloma, RR-=Relapsed and/or Refractory, ASCT=Autologous Stem Cell Transplantation, nCR=Near Complete Remission, NA=Not Available, CIV=Continuous Intravenous, PO=Oral, IV= Intravenous,

VRD has been compared with two other combinations in the phase 2 EVOLUTION trial.11 This randomized study concurrently evaluated VRD, bortezomib-cyclophosphamide-dexamethasone (VCD), and bortezomib-dexamethasone-cyclophosphamide-lenaldiomide (VDCR). VCD was later substituted with a modified VCD schedule which added an extra dose of cyclophosphamide on day 15. Initial treatment was followed by four six-week cycles of maintenance bortezomib in all arms. The best response was noted in the modified VCD regimen (PR or better in 100%) with nearly half of the patients achieving CR, although the number of patients enrolled in this arm was small (n=17). One-year OS was 92% for the 4-drug VDCR arm, and 100% for the other three regimens.11 The study shows that VCD is a reasonable, much less expensive alternative to VRD in terms of highly active 3 drug combinations. Further the study shows that the addition of cyclophosphamide to VRD (or lenalidomide to VCD) to creat a 4-drug combination does not increase efficacy, but results in higher toxicity rates. However, more studies are needed.

The ongoing phase III study (IFM/DFCI2009), designed to evaluate the clinical benefit from VRD, with or without immediate ASCT followed by lenalidomide maintenance, is an important trial attempting to address the usefulness of upfront transplantation in the era of such potent regimens (NCT01208662).

b) Thalidomide-Bortezomib

A recent phase 3 study compared bortezomib-thalidomide-dexamethasone (VTD) versus thalidomide-dexamethasone (TD) as pre-transplant induction therapy and post-double ASCT consolidation therapy (Table 4).12 The study showed higher CR/nCR rates in the VTD arm (31% vs. 11% with TD; p=0.0001). This improvement translated into superior progression free survival (PFS) rate (3-year PFS 68% vs.12%, respectively). No benefit was apparent in terms of OS.12 Despite exclusion of patients with ≥grade 2 neuropathy, the improvement in PFS came at a cost of significant neurotoxicity with VTD [10% grade 3 or higher PN similar to that in the Spanish trial (14%)13 using VTD induction], a significant deterrent to its long-term use. We feel that the use of this potent regimen of two neurotoxic agents is best reserved for clinical scenarios requiring immediate cytoreduction such as MM-induced acute renal failure. There are no randomized trials comparing VTD with either VCD or VRD. In most circumstances, including renal failure, we prefer VCD over VTD.

Table 4.

Phase 3 TRIALS WITH BORTEZOMIB (V)- BASED COMBINATIONS IN MULTIPLE MYELOMA*

Combination N Cohort ORR (%) PFS mo HR for PFS P value for PFS OS mo HR for OS P value for OS

1 GIMEMA12 NDMM ASCT eligible (3-yr) 0.63 0.006 (3-yr) NA 0.30
VTD X 3 +ASCT × 2+ VTD X 2 236 96 68% 86%
TD X 3+ ASCT × 2 + TD X 2 238 89 56% 84%

2 IFM 2007-0214 NDMM, ASCT eligible NA 0.22 NA NA NS
VD + ASCT 99 89 30
vtD+ASCT 100 86 26

3 PETHEMA19 NDMM ≥ 65 yrs 1.2 0.1 (3-yr) 1.2 0.3
VMP+ VT or VP maintenance 130 81 34 74%
VTP + VT or VP maintenance 130 80 25 65%

4 PETHEMA/GEM05MEN0S6568 NDMM <66 ASCT eligible NA NA 0.006 NA NS
TD 127 27 4-yr
VTD 130 NR 76%***
VBMCP/VBAD/B 129 38

5 VISTA16 NDMM ASCT ineligible 0.6 <0.001 0.7 <0.001
VMP 327 71 22 56
MP 328 35 15 43

6 UPFRONT22 NDDM Elderly, ASCT ineligible NA NS (1-yr) NA NS
VD+ V maintenance 168 73 14 87%
VTD+ V maintenance 167 80 15 86%
VMP+V maintenance 167 69 17 89%

7 HOVON 65/GMMG-HD431 NDMM +ASCT 3-yr 0.8 0.047 3-yr 0.7 .048
VAD with T maintenance 305 87 42% 71%
PAD with V maintenance 308 92 48% 78%

8 VMP NDMM ≥ 65 yrs ASCT Ineligible 0.6 0.008 3-yr 0.9 0.77
VMPT-VT20 257 81 27 89%
254 89 NR 87%

9 VANTAGE 08844 RR ** 0.8 0.01 0.9 0.35
Bortezomib+Vorinostat 347 56 8 NR
Bortezomib+placebo 320 41 7 28

10 IFM-EBMT69 RR after ASCT NA 0.001 (2-yr) NA 0.18
VTD 135 90 19 72%
TD 132 69 13 68%

11 DOXIL-MMY-300126 RR 0.6 <0.001 (1.25-yr) 0.7 0.03
V 322 41 9 65%
PLD+V 324 44 7 76%
*

Bortezomib-dexamethasone doublet excluded

**

Patients with prior BTZ resistance excluded

***

No OS difference in 3 arms

Abbreviations : ORR= Overall Response Rate, HR=Hazard Ratio, NDMM=Newly Diagnosed Multiple myeloma, ASCT, Autologous Stem Cell Transplantation, PFS=Progression-Free Survival, OS=Overall Survival, NA=not Available, NR=Not Reached

In order to improve upon the safety profile of VTD while preserving its marked efficacy (i.e. to design a regimen with best efficacy/toxicity ratio), the Intergroupe Francophone du Myelome (IFM) conducted a phase 3 trial comparing reduced dose bortezomib (1mg/m2 IV days 1, 4, 8 and 11 of a 3 week cycle), thalidomide (100mg/day), and dexamethasone (vtD) versus standard full-dose bortezomib-dexamethasone (VD).14 The dose of dexamethasone was similar in both the arms. An increment in the dosage of bortezomib and thalidomide was permitted if less than PR was achieved with 2 cycles of vtD. However, ninety percent of patients achieved a PR or better after just 2 cycles of vtD. The grade 3-4 peripheral neuropathy was 3% with vTD and 10% with VD. Although no difference in near CR or better responses were seen (31% vs. 22% with VD), a substantial increase in CR+VGPR rates was noted with vtD prior to ASCT (49% vs. 36% with VD). The PFS or OS outcomes between the 2 groups have not shown any difference so far.14 This study shows that optimizing the dose of bortezomib can alleviate the risk of neuropathy without sacrificing efficacy.

B. Combinations of bortezomib with conventional chemotherapeutic agents

The magnitude of synergy between conventional cytotoxic agents and bortezomib appears to be considerably high.15 Bortezomib-based combinations such as VCD and bortezomib, melphalan, prednisone (VMP) produce high response rates. However, in most trials, despite significant improvement in the CR rates (considered a surrogate for OS), an improvement in OS has not been seen.

a. Melphalan-Bortezomib based combinations

The landmark, international VISTA (Velcade as Initial Standard Therapy in Multiple Myeloma: Assessment with melphalan and prednisone) trial established the superiority of VMP (Bortezomib-melphalan and prednisone) compared with MP in transplant-ineligible patients with newly diagnosed myeloma.16,17 The VISTA trial (Table 4) found a 13-month improvement in the median OS with VMP compared with MP (56.4 versus 43.1 months respectively; HR 0.7, p=0.0004). However, the survival of patients with high-risk cytogenetics (n=46) did not improve with the addition of bortezomib, a reminder of the continued poor outcome in these patients despite several advances in myeloma therapy.17 No excess risk of second primary malignancies (SPM) was seen between the two arms.18

A subsequent Spanish study (Table 4) compared VMP with VTP (bortezomib, thalidomide and prednisone).19 This trial showed that the grade 3 or worse neurotoxicity of VMP seen in the VISTA trial (13%) could be substantially reduced (~5%) with modification of the bortezomib dose from twice per week to the once per week schedule.19 Similar observations of significantly reduced grade 3-4 non hematologic adverse effects, including peripheral neuropathy, without a compromise in efficacy were reported in an Italian study of 551 patients in which the schedule of bortezomib was modified from twice weekly to once weekly after enrollment of the first 139 patients (Table 4).20 A separate phase III trial conducted by the IFM demonstrated that the risk of neurotoxicity can be further reduced by subcutaneous dosing.21 Unless there is need for rapid control for myeloma (eg., acute renal failure, extramedullary disease, or plasma cell leukemia) we prefer once-weekly subcutaneous bortezomib as the standard schedule when using bortezomib in both newly diagnosed and relapsed settings.

The importance of using a modified bortezomib schedule to minimize neuropathy as much as possible was recently highlighted by the results of a recent community-based phase 3 trial of elderly patients that compared VD, VMP and VTD. This trial found that the overall average quality of life (QOL) of patients with myeloma failed to improve above baseline in any of the 3 arms after one year of therapy.22,23

b. Cyclophosphamide-Bortezomib based combinations

Alkylators are particularly effective in MM. Owing to the lack of cumulative stem cell damage, cyclophosphamide, unlike melphalan, preserves the ability to harvest stem cells during induction. The VCD regimen which combines cyclophosphamide and bortezomib is an excellent combination and has become our preferred bortezomib-based combination regimen in the frontline setting, as well as in the maintenance and relapsed settings for selected patients. VCD has shown high response rates (61% ≥VGPR and nearly 40% CR/nCR rates).24 A modified VCD regimen with weekly bortezomib and low-dose dex (40 mg weekly) is equally effective compared with the original VCD regimen that employed a twice weekly bortezomib schedule and high-dose dex, and is better tolerated.25 As suggested by the results of the EVOLUTION trial,11 VCD produces comparable results to VRD in newly diagnosed patients. Although the follow-up of patients in these trials is short to assess the survival outcomes, the regimens incorporating bortezomib and cyclophosphamide show rapid and deep responses and appear to be promising.

c. Anthracyclines-Bortezomib based combinations

Anthracyclines, doxorubicin and pegylated liposomal preparation (PLD) have been shown to enhance MM cell killing when combined with bortezomib in preclinical studies (Table 1). Although statistically significant benefit has been seen with the use of anthracyclines in myeloma, the true clinical benefits have been modest at best. PLD has improved pharmacokinetic and safety profile compared with doxorubicin, but is more expensive, and not easily available.

A phase III international study comparing bortezomib/PLD with bortezomib alone in relapsed, advanced MM, demonstrated improved response quality (27% CR+VGPR vs. 19% with bortezomib alone; p=0.015), response duration (10.2 months vs. 7 months with bortezomib alone, p=0.0008), PFS and OS (Table 4).26 The improved efficacy comes at a cost of greater toxicity (grade 3-4 myelosuppression, fatigue, diarrhea, hand-foot syndrome) although bortezomib induced neurotoxicity was not increased.26 Comparatively, a non-randomized study involving VDT ± liposomal doxorubicin (Table 2) showed further improvement in PFS with the four-drug regimen (15 vs. 8 months for VDT) again without any increment in the treatment-emergent neuropathy rate in the doxorubicin containing arm compared to VDT alone. In addition this study registered responses even in patients who were previously anthracycline refractory. The cycle length in the latter study was 4 weeks instead of 3 weeks with bortezomib+ PLD phase III study.27

In the frontline setting, 3 published phase 2 studies have reported on the efficacy and tolerability of bortezomib combined with anthracyclines (Table 2). The recently reported outcome with intravenous DVD regimen demonstrated the advantages of using modified metronomic dosing of PLD (5mg/m2 along with dexamethasone 40mg and bortezomib 1 mg/m2 days1, 4, 8 and 11) in a longer four week schedule.28 This modified regimen retained the high efficacy of a previously reported study of same combination by Jakubowiak et al.29 with marked improvement in the side-effect profile (reduced fatigue, PN myelosuppression and palmar-plantar erythrodysesthesia).28 Similarly, Popat et al reported that bortezomib dose reduction from 1.3 mg/m2 to 1.0 mg/m2 improved the toxicity profile of this highly active regimen without adversely affecting the outcome measures.30 A randomized phase 3 HOVON65/GMMG-HD4 trial (Table 4) compared PAD with VAD (vincristine, Adriamycin and dexamethasone) induction regimen followed high-dose melphalan (HDM) therapy (in both arms) and post-transplant maintenance therapy bortezomib (in PAD arm) or thalidomide (VAD arm).31,32 Preliminary results suggest that PAD results in a higher ORR compared with VAD (80% for PAD vs. 64% post induction) and that this difference is sustained post HDM (92% vs. 87% for VAD; p=0.01). Out of the seven phase 3 trials of pre-transplant induction therapy the HOVON65/GMMG-HD4 trial is the only one to show survival advantage of a bortezomib-based regimen.31 However, the possible unavailability of bortezomib-based salvage therapy at relapse for all patients in the non-bortezomib VAD arm in this study could account for the survival difference. The investigators also analyzed the prognostic value of chromosomal abnormalities in a subgroup of 354 MM patients. Importantly, the patients with del 17p13 appeared to benefit the most from the bortezomib-containing regimen: the median PFS in VAD arm was 12.0 months vs. 26.2 months in PAD (P = .024); the 3 year-OS for arm VAD was 17% versus was 69% (P = .028) for PAD. After multivariate analysis, del17p13 was an independent predictor for PFS (P < .0001) and OS (P < .0001) in VAD arm, whereas no statistically significant effect on PFS (P = .28) or OS (P = .12) was seen with PAD 33.

d. VDT-PACE

The multi-agent chemotherapy regimen VDT-PACE (bortezomib, high dose dexamethasone, thalidomide, cisplatin, doxorubicin (adriamycin), cyclophosphamide and etoposide) has been tested as the standard induction therapy in the total therapy 3 (TT3) trials conducted by the Myeloma Institute for Research and Therapy in Arkansas It is an extremely potent combination that builds by combining the backbone of VTD therapy and the four-day continuous infusion of chemotherapy developed using the components of CAD and DCEP regimens utilized in earlier studies.34 With similar baseline prognostic features, event-free survival (EFS) and CR duration were markedly superior with TT3 vs. total therapy 2 (TT2) (Table 2), regardless of the addition of thalidomide to one arm of TT2.34 A recently updated analysis predicted a 5-year OS of 86% (CI 82.2-89.9) in patients with low-risk gene expression profile (GEP), no cytogenetic abnormalities, β2M ≤5.5 mg/L and LDH < 190 U/L vs. only 11.18% (CI 10.15-12.31) with high-risk GEP, cytogenetic abnormalities, β2M > 5.5 mg/L and LDH ≥190 U/L, highlighting the poor prognosis of the very high-risk group despite aggressive treatment.35 A recent trial (2006-66) replacing VDT/TD consolidation/maintenance during TT3 by VRD for 3 years demonstrated similar results (Table 2).36

Patients with t(4;14) on TT3 protocol had similar OS compared to the standard-risk patients, underscoring the efficacy of bortezomib-based combination therapy and ASCT in this high-risk category. The GIMEMA and HOVON-65/GMMG-HD4 protocols utilizing bortezomib with VTD induction plus post-ASCT consolidation and PAD, respectively also confirmed this effect seen with the TT3 protocol on patients harboring t(4;14).

We use VDT-PACE in newly diagnosed patients presenting with agrressive extramedullary disease or plasma cell leukemia, and in selected patients with relapsed refractory myeloma.

C. Experimental Combinations

The combinations listed here have some preclinical rationale. However, most if not all of the agents that are being tested in combination with bortezomib have failed to show significant single-agent activity in myeloma in phase 1/2 studies which is disappointing. The proof of the efficacy of these combinations will need to come from well controlled phase 3 studies. Such data are largely lacking at this juncture. Nevertheless we will review the current status of these combinations in detail since they may provide insight into directions for the future.

a.Bortezomib and Heat Shock Protein Inhibitors (HSPI)

HSP90 is a molecular chaperone for several oncoproteins that are crucial for the cancer cell growth, survival and drug resistance. Preclinical studies have demonstrated marked antitumor effects of HSP90 inhibitors even in MM, a malignancy which is not critically dependent upon the HSP client proteins.37 The upregulation of HSPs (for example, HSP90) by bortezomib is a cytoprotective response that leads to resistance to bortezomib. MM cells appear to be exquisitely sensitive to dual inhibition of the proteasome and HSP90. The affinity of HSP90 inhibitor, tanespimycin to tumor-related HSP90 is 100-fold higher compared to HSP from normal cells, making it an attractive therapeutic target. The early reports of clinical efficacy of HSP90 inhibitor-bortezomib combination are shown in Table 3. Interestingly, low rates of neutropenia, anorexia and peripheral neuropathy (PN) compared to historical data with bortezomib monotherapy were noted, and hepatotoxicity was manageable. The low rates of severe PN may be related to the neuroprotective effect of tanespimycin.38 The TIME-2 randomized study comparing bortezomib plus three different doses of tanespimycin in relapsed-refractory MM patients was truncated early for non-scientific reasons. Prior use of lenalidomide and bortezomib was a prerequisite for eligibility in this study. The preliminary results suggest an overall response rate (ORR) of 14% in this heavily pretreated (median number of therapies=5) population (Table 3).39 A phase 1/2 trial evaluating another novel HSP inhibitor, KW 2478 in combination with bortezomib is currently ongoing (NCT01063907). The results of a completed phase 3, open-label trial (NCT00546780). for patients with MM in first relapse, comparing tanespimycin plus fixed-dose of bortezomib with bortezomib alone would hopefully shed light on the efficacy of this doublet which has demonstrated modest activity to date.

Table 3.

Selected Phase 1 and 2 Trials of Combinations involving Bortezomib (V) and Enzyme Inhibitors or Monoclonal Antibodies in Relapsed and/or Refractory Multiple Myeloma

Class/ Agent(s) Phase N CBR % CR % PR % SD % PFS mo OS mo Comments

Heat Shock Protein 90 Inhibitor
i) Tanespimycin + V MTD: 340 mg/m2 IV38 1/2 72 27 3 12 33 NA 18 Severe PN not noted possibly due to neuroprotective effects of tanespimycin Lower anorexia, constipation & neutropenia rates compared to V alone.
V naïve 48 7.2 NR
V pretreated 22 3.7 17.7
V refractory 13 1.6 13.8

TIME 2 Tanespimycin+V39 2 22 14 0 9 45 NA NA Study prematurely terminated due to resource-related reasons

Deacetylase Inhibitors
i) VANTAGE 09565 Vorinostat+V ± Dex after 4 cycles if SD 2b 143 31 1 16 46 3 11 Activity shown in a heavily pre- treated cohort.(median 4 lines); V refractory and refractory, intolerant, or ineligible for IMiD-based therapy

ii) PANORAMA-241 anabinostat+V+Dex 2 55** 49 0 20 NA NA NA A relatively low rate of PN (24%) seen. 2% grade 3/4 PN observed. contrast romidepsin+VD

iii) Panabinostat+V ± Dex (San Miguel) 238 1B Escalation part 47 76 13 40 15 NA NA Common GD3/4 AEs thrombocytopenia, neutropenia, asthenia and anemia
Expansion part 15
V refractory 66

iv) Romidepsin +V+ Dex42 1/2 25 72 8 52 8 7* 76% at 1-yr PN rates (76%) almost twice that reported in APEX (36%).

AKT/mTOR/Farnesyl Transferase or Serine/Threonine Kinase Inhibitors
i) Perifosine+V± Dex50 1/2 84 41 4 18 41 6 25 Overall well tolerated; side effects, including GI toxicities and fatigue were manageable.
V relapsed :20 65 10 35 35 9 NR
V refractory: 53 32 2 11 43 6 23

ii) Temsirolimus+V48 1 20 20 0 5 60 6 11 3 deaths noted during treatment. Cytopenias commonly seen
2 43 47 5 19 44 5 NR

iii) Tipifarnib+V52 1 16 13 NA 32 NA NA Commonest AE Gd 2 diarrhea (24%)

iv) Enzastaurin+V66 1 23 NA 0 17 39 8* NA Commonest AE anemia (70%)

Monoclonal Antibodies
i) Elotuzumab (E)+V55 1 28 63 7 41 NA 10* NA E related key toxicities: infusion reactions

ii) Siltuximab+V67 2 21 NA 14 43 NA NA NA 33% discontinued therapy due to AE

Abbreviations : CBR= Clinical Benefit Rate, CR=Complete Remission, PR=Partial Remission, SD=Stable Disease, PFS=Progression-free survival, OS=Overall Survival, NA=Not Available, NR=Not Reached, Gd =Grade, PN=Peripheral Neuropathy, AE=Adverse Effects, IMiDs=Immunomodulatory Drugs,

b. Deacetylase Inhibitors (DACIs) with Bortezomib

In-vitro data suggest that DACIs exhibit potent anti-myeloma activity in combination with bortezomib by inhibiting aggresome formation, or by enhancing the histone acetylation properties of bortezomib (Table 1). Deacetylases (DACs) are enzymes that remove the acetyl groups from several target (both histone and non-histone) proteins including HSP90, HIF-1a, BCL-6, p53, involved in the DNA repair, gene expression and cell growth. DACIs reverse the deacetylated status of MM cells and lead to persistent acetylation. This promotes expression of genes that induce cell cycle arrest and apoptosis.40 Four clinical trials evaluating the efficacy of DACI in MM as monotherapy have demonstrated modest activity, but acceptable safety profile. The PANORAMA 2 study, a multicenter, phase 2 trial of pan deacetylase inhibitor, panabinostat-bortezomib and dexamethasone in patients who had to be bortezomib-refractory for enrollment showed an ORR of nearly 25%.41 Another recently reported small phase1/2 study evaluating the combination of bortezomib, dexamethasone and romidepsin (an HDAC inhibitor, currently approved for cutaneous T cell lymphoma) in the relapsed-refractory MM demonstrated a 60% PR rate.42 A retrospective review of nine patients, refractory to VRD who received vorinostat (Z) additionally (VRDZ) demonstrated 89% disease-control rate (stable disease or better) but disappointingly short median duration of response of 3 months and median OS of 4 (3-21) months.43

The above results with Bortezomib-DACI combinations (Table 3) while interesting need confirmation in phase 3 trials to isolate what benefit, if any, is due to the addition of DACI. Two phase III trials: the VANTAGE 088 study44 (a randomized, double blind placebo controlled phase 3 trial of bortezomib versus vorinostat plus bortezomib) and PANORAMA 1 trial45 (panabinostat, bortezomib versus bortezomib) are addressing this. Prelimininary results of VANTAGE 088 have been reported and are sobering, with no clinically significant improvement in PFS (the median improvement was 26 days), and no improvement in OS (Table 4). The results of PANORAMA 1 trial are awaited.16

c. AKT and mTOR Inhibitors plus Bortezomib

The activated PI3-K/AKT/mTOR pathway is a critical cytokine stimulated pro-tumoral pathway46 that mediates growth and survival of MM cells through their interaction with the BMSCs. TORC1 complex of the downstream mTOR kinase is inhibited by rapalogs. However, TORC1 inhibition-associated feedback activation of the PI3K/AKT pathway protects against apoptosis, and TORC1 inhibitors can only induce cell-cycle arrest without apoptosis. Dual TORC1/2 complex inhibitors may overcome the activated AKT mediated resistance seen with use of TORC1 inhibitors alone. In fact, a recent preclinical study has shown that TORC1/2 knock-down significantly inhibits the proliferative capacity of MM cells.47 However the issue of the feedback activation of the MEK/ERK resistance pathway as a result of TORC1 inhibition still remains. Therefore, specific inhibitors of TORC2 alone that inhibit phosphorylation and activation of AKT and are currently under development could potentially be more useful.

The postulation of synergy between PI3K/AKT/mTOR pathway inhibition and bortezomib formed the basis of a recent phase1/2 trial studying the steroid sparing regimen of weekly temsirolimus (a rapalog with anti-TORC1 activity) and bortezomib in heavily-pretreated relapsed and refractory MM patients. In the phase 2 portion, a third of patients (14 out of 43) attained PR or better. Myelosuppression, particularly thrombocytopenia was the most common toxicity observed.48

Resistance to bortezomib-induced apoptosis has been demonstrated to be associated with AKT, β-catenin and survivin (a member of inhibitor of apoptosis family) upregulation. Perifosine is a novel, proapoptotic, signal transduction modulator which directly inhibits the constitutively phosphorylated AKT in MM cells. This well-tolerated, oral agent has demonstrated activity in relapsed/refractory MM in combination with both the conventional and novel anti-myeloma agents.49 Recently, a phase 1/2 study of perifosine in combination with bortezomib with or without dexamethasone evaluated a heavily pretreated cohort of 84 patients with relapsed and /or refractory MM (Table 3).50 An ORR of 41% (65% in bortezomib–relapsed and 32% in bortezomib refractory) was noted. GI toxicities, fatigue and musculoskeletal pain were the predominant toxicities encountered. A phase III multicenter trial of testing the value of perifosine in relapsed/refractory patients previously treated with bortezomib is ongoing (NCT01002248).

d. Farnesyl transferase inhibitors (FTIs)

A phase 2 trial evaluating the efficacy of single agent FTI, tipifarnib was conducted on the premise that MM patients with mutated Ras exhibit reduced chemosensitivity and targeting farnesylation of oncoprotein Ras would inhibit its membrane association and signaling activity.51 The study involved forty-three heavily pre-treated patients with advanced MM and demonstrated disease stabilization in 64% of patients independent of farnesyl inhibition. The drug was well tolerated (toxicities included fatigue, diarrhea, myelosuppression and neuropathy) and 40 patients with SD remained stable for 5 months or more.51 This study along with the demonstration of pre-clinical synergistic activity of combination therapy (Table 1) served as the basis for phase I trial evaluating combination of bortezomib and FTI in relapsed/refractory MM (Table 3). Again, so far only stabilization of disease has been reported in nearly 50% of patients and the optimal dose is yet to be defined.52 The sequence of drug administration in the combination is important and bortezomib followed by FTI is recommended for optimal efficacy.52 The combination appears to be effective, but more data are required.

e. Monoclonal Antibodies (MAb)

Potential targets for MAbs in MM include antigens predominantly expressed on MM cells, growth factors, their receptors and signaling proteins although to date only modest clinical activity has been observed with MAb monotherapy in MM.53 While meaningful responses have been noted with well tolerated bortezomib-based regimens incorporating elotuzumab (chimeric anti-IL6 monoclonal antibody) or siltuximab, a chimeric anti-IL6 monoclonal antibody, (Table 3), the results of a recent randomized phase 2 trial of bortezomib vs. bortezomib+ mapatumumab (a fully human agonistic antibody activating the tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) receptor-1) in relapsed refractory patients were largely disappointing.54 A phase 2 trial of MPV with or without siltuximab is currently underway (NCT00911859).

Out of the many MAb combination therapies evaluated so far, elotuzumab-based combinations with novel agents, bortezomib or lenalidomide appear to be the most promising in relapsed refractory MM. In a phase 1 study of bortezomib and elotuzumab nearly two-thirds of patients achieved a clinically meaningful response (≥MR) with nearly one-half of all evaluable patients achieving at least PR.55 The median time to progression was 9.5 months. Infusion-related toxicities and grade 3-4 lymphopenia (25%) were commonly encountered, although adverse effects were largely manageable.55

The place of MAb therapy in the current treatment paradigms for MM is still evolving. The results of ongoing trials with MAbs plus existing novel agent-based therapies will define the roles of these agents that have a relatively favorable toxicity profile.

Conclusions and Future Directions

Implicit in our review is the development of numerous potent bortezomib-based combinations that have spawned during the past decade. Importantly, the economic implications of combining expensive agents deserve to be factored into the clinical decision making. The UPFRONT22 and the PETHEMA trials19 comparing the efficacy of different bortezomib-based combinations have been unable to establish the superiority of one specific combination. Specifically, the high-risk patients continue to have a poor outcome, although some bortezomib-based regimens appear to be able to overcome the adverse prognostic effects of t(4;14) when bortezomib-based induction and maintenance therapy is incorporated into the treatment plan, particularly in the context of tandem transplantation. With time, neoplastic plasma cells acquire a succession of distinctive features that enable them to proliferate via activation of alternative pathways notwithstanding the plethora of available combination therapies. Several next-generation PIs with molecular characteristics different from bortezomib are being evaluated, and a few that target different proteasome subunits can somewhat overcome bortezomib resistance when used alone or with other effective agents.

While several combinations outlined in this review are frequently being utilized, ongoing trials allow one to envision an optimized approach to proteasome inhibition that incorporates rational use of bortezomib as well as newer second-generation PIs such as carfilzomib, marizomib, and MLN 9708. Desirable properties such as oral route of administration and improved safety profile may make some PIs (e.g., MLN 9708) attractive agents if clinical efficacy is confirmed. The last decade has established a solid foundation upon which many rational bortezomib- and the next-generation PI-based combination regimens can be developed to further improve the outcome of patients afflicted with multiple myeloma.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Rajkumar SV, Richardson PG, Hideshima T, Anderson KC. Proteasome inhibition as a novel therapeutic target in human cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005;23(3):630–639. doi: 10.1200/JCO.2005.11.030. Prepublished on 2005/01/22 as. [DOI] [PubMed] [Google Scholar]
  • 2.Richardson PG, Barlogie B, Berenson J, et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. The New England journal of medicine. 2003;348(26):2609–2617. doi: 10.1056/NEJMoa030288. Prepublished on 2003/06/27 as. [DOI] [PubMed] [Google Scholar]
  • 3.Kumar SK, Lee JH, Lahuerta JJ, et al. Risk of progression and survival in multiple myeloma relapsing after therapy with IMiDs and bortezomib: A multicenter international myeloma working group study. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2012;26(1):149–157. doi: 10.1038/leu.2011.196. Prepublished on 2011/07/30 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Orlowski RZ, Stinchcombe TE, Mitchell BS, et al. Phase I trial of the proteasome inhibitor PS-341 in patients with refractory hematologic malignancies. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2002;20(22):4420–4427. doi: 10.1200/JCO.2002.01.133. Prepublished on 2002/11/15 as. [DOI] [PubMed] [Google Scholar]
  • 5.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;106(6):1316–1319. doi: 10.1002/cncr.21740. Prepublished on 2006/02/14 as. [DOI] [PubMed] [Google Scholar]
  • 6.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. British journal of haematology. 2008;143(4):537–540. doi: 10.1111/j.1365-2141.2008.07359.x. Prepublished on 2008/09/12 as. [DOI] [PubMed] [Google Scholar]
  • 7.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;110(10):3557–3560. doi: 10.1182/blood-2006-08-036947. Prepublished on 2007/08/11 as. [DOI] [PubMed] [Google Scholar]
  • 8.Ling SC, Lau EK, Al-Shabeeb A, et al. Response of myeloma to proteasome inhibitor bortezomib is correlated with unfolded protein response regulator XBP-1. Haematologica. 2011 doi: 10.3324/haematol.2011.043331. Prepublished on 2011/10/14 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Richardson PG, Weller E, Lonial S, et al. Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma. Blood. 2010;116(5):679–686. doi: 10.1182/blood-2010-02-268862. Prepublished on 2010/04/14 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Anderson KC, Jagannath S, Jakubowiak A, et al. Lenalidomide, bortezomib, and dexamethasone in relapsed/refractory multiple myeloma (MM): Encouraging outcomes and tolerability in a phase II study. ASCO Meeting; 2009. p. 8536. Abstracts. [Google Scholar]
  • 11.Kumar S, Flinn IW, Richardson PG, et al. Novel Three- and Four-Drug Combination Regimens of Bortezomib, Dexamethasone, Cyclophosphamide, and Lenalidomide, for Previously Untreated Multiple Myeloma: Results From the Multi-Center, Randomized, Phase 2 EVOLUTION Study. ASH Annual Meeting; 2010. p. 621. Abstracts. [Google Scholar]
  • 12.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;376(9758):2075–2085. doi: 10.1016/s0140-6736(10)61424-9. Prepublished on 2010/12/15 as. [DOI] [PubMed] [Google Scholar]
  • 13.Rosinol L, Cibeira MT, Martinez J, et al. Thalidomide / Dexamethasone (TD) Vs. Bortezomib (Velcade)a/Thalidomide / Dexamethasone (VTD) Vs. VBMCP/VBAD/Velcadea as Induction Regimens Prior Autologous Stem Cell Transplantation (ASCT) in Multiple Myeloma (MM): Results of a Phase III PETHEMA/GEM Trial. ASH Annual Meeting; 2009. p. 130. Abstracts. [Google Scholar]
  • 14.Moreau P, Avet-Loiseau H, Facon T, et al. Bortezomib plus dexamethasone versus reduced-dose bortezomib, thalidomide plus dexamethasone as induction treatment prior to autologous stem cell transplantation in newly diagnosed multiple myeloma. Blood. 2011 doi: 10.1182/blood-2011-05-355081. Prepublished on 2011/08/19 as. [DOI] [PubMed] [Google Scholar]
  • 15.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;101(6):2377–2380. doi: 10.1182/blood-2002-06-1768. Prepublished on 2002/11/09 as. [DOI] [PubMed] [Google Scholar]
  • 16.San Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. The New England journal of medicine. 2008;359(9):906–917. doi: 10.1056/NEJMoa0801479. Prepublished on 2008/08/30 as. [DOI] [PubMed] [Google Scholar]
  • 17.Miguel JFS, 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. ASH Annual Meeting; 2011. p. 476. Abstracts. [Google Scholar]
  • 18.Miguel JFS, Richardson PG, Orlowski RZ, et al. Risk of Second Primary Malignancies (SPMs) Following Bortezomib (Btz)-Based Therapy: Analysis of Four Phase 3 Randomized Controlled Trials in Previously Untreated or Relapsed Multiple Myeloma (MM). ASH Annual Meeting; 2011. p. 2933. Abstracts. [Google Scholar]
  • 19.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;11(10):934–941. doi: 10.1016/s1470-2045(10)70187-x. Prepublished on 2010/08/27 as. [DOI] [PubMed] [Google Scholar]
  • 20.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;28(34):5101–5109. doi: 10.1200/jco.2010.29.8216. Prepublished on 2010/10/14 as. [DOI] [PubMed] [Google Scholar]
  • 21.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;12(5):431–440. doi: 10.1016/s1470-2045(11)70081-x. Prepublished on 2011/04/22 as. [DOI] [PubMed] [Google Scholar]
  • 22.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. ASH Annual Meeting; 2011. p. 478. Abstracts. [Google Scholar]
  • 23.Niesvizky R, Flinn IW, Rifkin R, et al. Patient-Reported Quality of Life (QoL) in Elderly, Newly Diagnosed Multiple Myeloma (MM) Patients Receiving Bortezomib-Based Combinations: Results From All Randomized Patients in the Community-Based, Phase 3b UPFRONT Study. ASH Annual Meeting; 2011. p. 1864. Abstracts. [Google Scholar]
  • 24.Reeder CB, Reece DE, Kukreti V, et al. Cyclophosphamide, bortezomib and dexamethasone induction for newly diagnosed multiple myeloma: high response rates in a phase II clinical trial. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2009;23(7):1337–1341. doi: 10.1038/leu.2009.26. Prepublished on 2009/02/20 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Reeder CB, Reece DE, Kukreti V, et al. Once- versus twice-weekly bortezomib induction therapy with CyBorD in newly diagnosed multiple myeloma. Blood. 2010;115(16):3416–3417. doi: 10.1182/blood-2010-02-271676. Prepublished on 2010/04/24 as. [DOI] [PubMed] [Google Scholar]
  • 26.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. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007;25(25):3892–3901. doi: 10.1200/JCO.2006.10.5460. Prepublished on 2007/08/08 as. [DOI] [PubMed] [Google Scholar]
  • 27.Ciolli S, Leoni F, Casini C, Breschi C, Santini V, Bosi A. The addition of liposomal doxorubicin to bortezomib, thalidomide and dexamethasone significantly improves clinical outcome of advanced multiple myeloma. British journal of haematology. 2008;141(6):814–819. doi: 10.1111/j.1365-2141.2008.07147.x. Prepublished on 2008/04/16 as. [DOI] [PubMed] [Google Scholar]
  • 28.Berenson JR, Yellin O, Chen CS, et al. A modified regimen of pegylated liposomal doxorubicin, bortezomib and dexamethasone (DVD) is effective and well tolerated for previously untreated multiple myeloma patients. Br J Haematol. 2011 doi: 10.1111/j.1365-2141.2011.08884.x. Prepublished on 2011/09/29 as. [DOI] [PubMed] [Google Scholar]
  • 29.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. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27(30):5015–5022. doi: 10.1200/JCO.2008.19.5370. Prepublished on 2009/09/10 as. [DOI] [PubMed] [Google Scholar]
  • 30.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. British journal of haematology. 2008;141(4):512–516. doi: 10.1111/j.1365-2141.2008.06997.x. Prepublished on 2008/03/29 as. [DOI] [PubMed] [Google Scholar]
  • 31.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). ASH Annual Meeting; 2010. p. 40. Abstracts. [Google Scholar]
  • 32.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). ASH Annual Meeting; 2008. p. 653. Abstracts. [Google Scholar]
  • 33.Neben K, Lokhorst HM, Jauch A, et al. Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p. Blood. 2012;119(4):940–948. doi: 10.1182/blood-2011-09-379164. [DOI] [PubMed] [Google Scholar]
  • 34.Pineda-Roman M, Zangari M, Haessler J, et al. Sustained complete remissions in multiple myeloma linked to bortezomib in total therapy 3: comparison with total therapy 2. British journal of haematology. 2008;140(6):625–634. doi: 10.1111/j.1365-2141.2007.06921.x. Prepublished on 2008/02/28 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hoering A, Mitchell A, Sexton R, Crowley J, Barlogie B. Prognostic Index for Predicting Overall Survival and Event-Free Survival In Total Therapy 3 Patients. ASH Annual Meeting; 2010. p. 1342. Abstracts. [Google Scholar]
  • 36.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(21):4168–4173. doi: 10.1182/blood-2009-11-255620. Prepublished on 2010/02/04 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Mitsiades CS, Mitsiades NS, McMullan CJ, et al. Antimyeloma activity of heat shock protein-90 inhibition. Blood. 2006;107(3):1092–1100. doi: 10.1182/blood-2005-03-1158. Prepublished on 2005/10/20 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Richardson PG, Chanan-Khan AA, Lonial S, et al. Tanespimycin and bortezomib combination treatment in patients with relapsed or relapsed and refractory multiple myeloma: results of a phase 1/2 study. Br J Haematol. 2011;153(6):729–740. doi: 10.1111/j.1365-2141.2011.08664.x. Prepublished on 2011/05/04 as. [DOI] [PubMed] [Google Scholar]
  • 39.Richardson PG, Badros AZ, Jagannath S, et al. Tanespimycin with bortezomib: activity in relapsed/refractory patients with multiple myeloma. Br J Haematol. 2010;150(4):428–437. doi: 10.1111/j.1365-2141.2010.08264.x. Prepublished on 2010/07/14 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.McConkey D. Proteasome and HDAC: who’s zooming who? Blood. 2010;116(3):308–309. doi: 10.1182/blood-2010-04-278507. Prepublished on 2010/07/24 as. [DOI] [PubMed] [Google Scholar]
  • 41.Richardson PG, Alsina M, Weber DM, et al. Phase II Study of the Pan-Deacetylase Inhibitor Panobinostat in Combination with Bortezomib and Dexamethasone in Relapsed and Bortezomib-Refractory Multiple Myeloma (PANORAMA 2). ASH Annual Meeting; 2011. p. 814. Abstracts. [Google Scholar]
  • 42.Harrison SJ, Quach H, Link E, et al. A high rate of durable responses with romidepsin, bortezomib, and dexamethasone in relapsed or refractory multiple myeloma. Blood. 2011;118(24):6274–6283. doi: 10.1182/blood-2011-03-339879. Prepublished on 2011/09/14 as. [DOI] [PubMed] [Google Scholar]
  • 43.Siegel D, B E, McBride L, Richarson P, Schmidt L, Gao Z, Tufail M, Anand P. Vorinostat overcomes resistance in patients with multiple myeloma refractory to bortezomib, lenalidonide and dexamethasone. Hematologica. 2011;2011(96(s1)):S94. [Google Scholar]
  • 44.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. ASH Annual Meeting; 2011. p. 811. Abstracts. [Google Scholar]
  • 45.San-Miguel JF, de Moraes Hungria VT, Yoon S-S, et al. Update on a Phase III Study of Panobinostat with Bortezomib and Dexamethasone in Patients with Relapsed Multiple Myeloma: PANORAMA 1. ASH Annual Meeting; 2011. p. 3976. Abstracts. [Google Scholar]
  • 46.Hidalgo M. From node to pathway blockade: lessons learned from targeting mammalian target of rapamycin. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30(1):85–87. doi: 10.1200/JCO.2011.37.9701. Prepublished on 2011/11/10 as. [DOI] [PubMed] [Google Scholar]
  • 47.Maiso P, Liu Y, Morgan B, et al. Defining the role of TORC1/2 in multiple myeloma. Blood. 2011;118(26):6860–6870. doi: 10.1182/blood-2011-03-342394. Prepublished on 2011/11/03 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ghobrial IM, Weller E, Vij R, et al. Weekly bortezomib in combination with temsirolimus in relapsed or relapsed and refractory multiple myeloma: a multicentre, phase 1/2, open-label, dose-escalation study. Lancet Oncol. 2011;12(3):263–272. doi: 10.1016/s1470-2045(11)70028-6. Prepublished on 2011/02/25 as. [DOI] [PubMed] [Google Scholar]
  • 49.Hideshima T, Catley L, Yasui H, et al. Perifosine, an oral bioactive novel alkylphospholipid, inhibits Akt and induces in vitro and in vivo cytotoxicity in human multiple myeloma cells. Blood. 2006;107(10):4053–4062. doi: 10.1182/blood-2005-08-3434. Prepublished on 2006/01/19 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Richardson PG, Wolf J, Jakubowiak A, et al. Perifosine plus bortezomib and dexamethasone in patients with relapsed/refractory multiple myeloma previously treated with bortezomib: results of a multicenter phase I/II trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2011;29(32):4243–4249. doi: 10.1200/JCO.2010.33.9788. Prepublished on 2011/10/13 as. [DOI] [PubMed] [Google Scholar]
  • 51.Alsina M, Fonseca R, Wilson EF, et al. Farnesyltransferase inhibitor tipifarnib is well tolerated, induces stabilization of disease, and inhibits farnesylation and oncogenic/tumor survival pathways in patients with advanced multiple myeloma. Blood. 2004;103(9):3271–3277. doi: 10.1182/blood-2003-08-2764. Prepublished on 2004/01/17 as. [DOI] [PubMed] [Google Scholar]
  • 52.Lonial S, Francis D, Karanes C, et al. A Phase I MMRC Clinical Trial Testing the Combination of Bortezomib and Tipifarnib in Relapsed/Refractory Multiple Myeloma. ASH Annual Meeting; 2008. p. 3706. Abstracts. [Google Scholar]
  • 53.Richardson PG, Lonial S, Jakubowiak AJ, Harousseau JL, Anderson KC. Monoclonal antibodies in the treatment of multiple myeloma. Br J Haematol. 2011 doi: 10.1111/j.1365-2141.2011.08790.x. Prepublished on 2011/07/23 as. [DOI] [PubMed] [Google Scholar]
  • 54.Belch A, Sharma A, Spencer A, et al. A Multicenter Randomized Phase II Trial of Mapatumumab, a TRAIL-R1 Agonist Monoclonal Antibody, In Combination with Bortezomib In Patients with Relapsed/Refractory Multiple Myeloma (MM). ASH Annual Meeting; 2010. p. 5031. Abstracts. [Google Scholar]
  • 55.Jakubowiak AJ, Benson DM, Jr, Bensinger W, et al. Elotuzumab In Combination with Bortezomib In Patients with Relapsed/Refractory Multiple Myeloma: Updated Results of a Phase 1 Study. ASH Annual Meeting; 2010. p. 3023. Abstracts. [Google Scholar]
  • 56.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(7):1350–1356. doi: 10.1038/leu.2010.116. Prepublished on 2010/05/29 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jakubowiak AJ, Griffith KA, Reece DE, et al. Lenalidomide, bortezomib, pegylated liposomal doxorubicin, and dexamethasone in newly diagnosed multiple myeloma: a phase 1/2 Multiple Myeloma Research Consortium trial. Blood. 2011;118(3):535–543. doi: 10.1182/blood-2011-02-334755. Prepublished on 2011/05/21 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kropff M, Liebisch P, Knop S, et al. DSMM XI study: dose definition for intravenous cyclophosphamide in combination with bortezomib/dexamethasone for remission induction in patients with newly diagnosed myeloma. Annals of hematology. 2009;88(11):1125–1130. doi: 10.1007/s00277-009-0726-6. Prepublished on 2009/03/11 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sher T, Ailawadhi S, Miller KC, et al. A steroid-independent regimen of bortezomib, liposomal doxorubicin and thalidomide demonstrate high response rates in newly diagnosed multiple myeloma patients. Br J Haematol. 2011;154(1):104–110. doi: 10.1111/j.1365-2141.2011.08703.x. Prepublished on 2011/05/11 as. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Belch A, Reece DE, Bahlis NJ, et al. Bortezomib [VELCADETM], Pegylated Liposomal Doxorubicin [DOXIL/CAELYX(R)] and Dexamethasone in the Treatment of Previously Untreated Multiple Myeloma Patients: Impact on Quality-of-Life. ASH Annual Meeting. 2007;110(11):3618. Abstracts. [Google Scholar]
  • 61.Reece DE, Rodriguez GP, Chen C, et al. Phase I-II trial of bortezomib plus oral cyclophosphamide and prednisone in relapsed and refractory multiple myeloma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2008;26(29):4777–4783. doi: 10.1200/JCO.2007.14.2372. Prepublished on 2008/07/23 as. [DOI] [PubMed] [Google Scholar]
  • 62.Kropff M, Bisping G, Schuck E, et al. Bortezomib in combination with intermediate-dose dexamethasone and continuous low-dose oral cyclophosphamide for relapsed multiple myeloma. British journal of haematology. 2007;138(3):330–337. doi: 10.1111/j.1365-2141.2007.06656.x. Prepublished on 2007/07/07 as. [DOI] [PubMed] [Google Scholar]
  • 63.Palumbo A, Gay F, Bringhen S, et al. Bortezomib, doxorubicin and dexamethasone in advanced multiple myeloma. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO. 2008;19(6):1160–1165. doi: 10.1093/annonc/mdn018. Prepublished on 2008/03/11 as. [DOI] [PubMed] [Google Scholar]
  • 64.Berenson JR, Yellin O, Bessudo A, et al. Bendamustine Combined with Bortezomib Has Efficacy in Patients with Relapsed or Refractory Multiple Myeloma: A Phase 1/2 Study. ASH Annual Meeting. 2011;118(21):1857. Abstracts. [Google Scholar]
  • 65.Siegel DS, Dimopoulos MA, Yoon S-S, et al. Vantage 095: Vorinostat in Combination with Bortezomib in Salvage Multiple Myeloma Patients: Final Study Results of a Global Phase 2b Trial. ASH Annual Meeting. 2011;118(21):480. Abstracts. [Google Scholar]
  • 66.Ghobrial IM, Munshi NC, Harris BN, et al. A phase I safety study of enzastaurin plus bortezomib in the treatment of relapsed or refractory multiple myeloma. Am J Hematol. 2011;86(7):573–578. doi: 10.1002/ajh.22048. Prepublished on 2011/06/02 as. [DOI] [PubMed] [Google Scholar]
  • 67.Rossi J-F, Manges RF, Sutherland HJ, et al. Preliminary Results of CNTO 328, An Anti-Interleukin-6 Monoclonal Antibody, in Combination with Bortezomib in the Treatment of Relapsed or Refractory Multiple Myeloma. ASH Annual Meeting. 2008;112(11):867. Abstracts. [Google Scholar]
  • 68.Rosinol 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) Hematologica. 2011;2011(96(S1)):S69. [Google Scholar]
  • 69.Garderet L, I S, Moreau P, Dib M, Caillot D, Niederwieser D, Masszi T. Bortezomib (Velcade)-Thalidomide-Dexaemthasone (VTD) is superior to Thalidomide-Dexaemthasone (TD) in patients with multiple myeloma (MM) progressing or relapsing after autologous transplantation. Hematologica. 2011;2011(96(s1)):72–73. [Google Scholar]

RESOURCES