Open access

Induction Therapy in Multiple Myeloma

Written By

Sule Mine Bakanay and Meral Beksac

Submitted: 09 October 2012 Published: 10 April 2013

DOI: 10.5772/55151

From the Edited Volume

Multiple Myeloma - A Quick Reflection on the Fast Progress

Edited by Roman Hajek

Chapter metrics overview

1,860 Chapter Downloads

View Full Metrics

1. Introduction

Today, multiple myeloma (MM) can be defined as a heterogenous disease composed of different clinical conditions. The differences are a result of patient related factors (age, sex, comorbidity), disease related complications (renal failure, bone disease, neuropathy, thrombosis) and biological characteristics (cytogenetics, lactate dehydrogenase level, plasma cell labelling index, beta2-microglobulin, gene expression profiles). The widely used international scoring system is a powerful tool for determining survival. However, it cannot be used for treatment planning. The biological determinants of disease determined by flourescein in situ hybridization (FISH) and/or conventional cytogenetics are better tools to stratify myeloma subgroups with different survival profiles. Thus these are better tools for designing therapeutic approaches. A risk stratification of newly diagnosed MM according to FISH/Karyotyping has been recently reviewed by Rajkumar (Rajkumar, 2012).

High dose melphalan supported by autologous stem cell transplantation (ASCT) can increase response rates and prolong progression free and overal survival compared to conventional chemotherapies (Attal et al., 1996; Child et al., 2003; Fermand et al., 2005; Koreth et al., 2007). The initial induction regimen is chosen according to whether the patient is eligible or ineligible for a subsequent HDT-ASCT as well as the risk stratification of the patient. Advanced age or significant comorbidity are important limitations for ASCT. High dose therapy has been generally considered for patients ≤ 65 years. However, in medically fit patients, this can be extended up to age 70-75 years. Achievement of high quality responses (VGPR, CR/nCR) at the time of transplantation has been demostrated to be an early predictor of improved outcomes after ASCT (Harousseau et al., 2009; Chanan-Khan&Giralt, 2010). Elderly patients or patients ineligible for transplantation may also benefit from chemotherapy by achieving high quality responses preferably CR in terms of progression free survival (PFS) and overall survival (OS). As such, the choice of induction therapy is crucial for survival for most patients with MM. The emergence of novel agents (thalidomide, lenalidomide and bortezomib) and incorporation of these agents in to the current induction protocols has increased the rate of CR and at least VGPR before ASCT and significantly improved the OS in MM (Kumar et al., 2008; Kastritis et al., 2009). This opened a new area of debate ‘upfront’ versus ‘delayed’ transplantation. However, current recommendations from experts is that high dose therapy supported by autologous stem cell transplantation (HDT-ASCT) should be the standard of care for eligible patients (Ludwig et al., 2011).

Advertisement

2. Induction therapy for patients eligible for HDT–ASCT

In patients for whom HDT-ASCT is planned, the goal of induction treatment before HDT-ASCT should be to achieve the deepest response preferably up to the level of ≥ VGPR as quickly as possible, to reverse disease related complications and ameliorate patient’s symptoms. The protocol should not induce stem cell toxicity and impair stem cell collection. Hence, it is important to avoid melphalan prior to stem cell collection (Cavo et al., 2011; Ludwig et al., 2011).

Prior to novel agents, the standard of care for patients eligible for ASCT was based on high dose dexamethasone alone or VAD (Vincristine, adriamycin, high dose dexamethasone). Over the last few years, the emergence of novel agents (thalidomide, bortezomib and lenalidomide) has shifted the choice of induction regimen from conventional VAD or VAD-like regimens to novel-agent containing protocols. In the earlier studies, the combinations of novel agents with high dose dexamethasone were shown to be superior to VAD regimen before ASCT. The more recent trials have concentrated on upfront use of initially 2-drug and more recently 3-drug or even 4-drug combinations applied before ASCT.

2.1. Thalidomide–based regimens

Thalidomide is the first immunomodulatory drug used in the treatment of MM. Apart from the anti-angiogenic activity, this group of drugs also induce apoptosis in myeloma cells. Thalidomide induces responses in MM patients refractory to conventional and even high dose therapy suggesting that it can overcome drug resistance. It may alter the secretion and bioactivity of cytokines (e.g. TNF-α) secreted into bone marrow microenvironment by myeloma and bone marrow stromal cells that induce myeloma cell growth and survival. Thalidomide mediates its immunomodulatory action by induction of Th-1 T cell response with secretion of IFN-Y and IL-2 and regulation of adhesion molecule expression (Hideshima et al., 2000).

After the initial studies showing that single agent thalidomide could produce significant reponses and that combination of thalidomide with dexamethasone (TD) results in synergism in refractory/relapsed MM, thalidomide was introduced to the induction therapy for newly diagnosed MM (Singhal et al., 1999; Palumbo et al., 2001). The phase II studies demonstrated the efficacy of TD as front line therapy with 64%-66% response rates (Rajkumar et al., 2002; Cavo et al., 2004). In 2005, a retrospective matched case-control analysis provided the first demonstration of superior rate and depth of response by TD compared with VAD as induction ( ≥PR; 76% vs 52%, respectively, p<0.001) (Cavo et al., 2005). Based on the subsequent phase III studies which confirmed the superior response rates achieved with thalidomide containing regimens compared with the conventional induction therapies, TD regimen received an accelerated approval in patients with newly diagnosed MM (Rajkumar et al.,2006; Rajkumar et al., 2008). The summary of the phase II-III studies involving thalidomide-based induction regimens is shown in table-1.

Rajkumar et al. in a randomized double blind placebo-controlled study, provided the first data on significant prolongation of time to progression (TTP) and progression free survival (PFS) with TD compared with dexamethasone alone in patients with newly diagnosed MM ( 14.9 vs. 6.5 months; p<0.001). However, this study was not powered enough to compare the differences in OS (Rajkumar et al., 2008). Barlogie et al. randomized their patients to receive two cycles of high dose melphalan based chemotherapy each supported with ASCT (Total therapy-2) with or without thalidomide added from outset until disease progression and reported that addition of thalidomide improved the rate of CR and EFS but failed to prolong OS (Barlogie et al., 2006). In a retrospective pair-matched analysis, thalidomide incorporated to induction regimen and continued until the second ASCT revealed significantly improved clinical outcomes and a trend towards extended OS at 5 years ( 69% vs. 53%; p=0.07) (Cavo et al., 2009). The HOVON-50 trial incorporated doxorubicin to TD (TAD) and compared this regimen with conventional VAD as frontline therapy showing a better response before and after HDT-ASCT (Lokhorst et al., 2008). After long term follow-up, the TAD arm folowed by thalidomide maintenance after ASCT was able to induce longer event free survival compared to the VAD arm folloowed by interferon ( 34 vs. 22 months; p<0.001) but this did not translate into an improved OS (73 vs. 60 months; p=0.77). Which can be explained by a decreased survival from relapse while on thalidomide maintenance (Lokhorst et al., 2010). A recent MRC Myeloma IX randomized trial compared oral combination therapy CTD (cyclophosphamide, thalidomide, dexamethasone) with oral cyclophosphamide incorporated into conventional VAD (CVAD). Significantly superior response rates were attained with CTD compared with CVAD both after induction and after ASCT. CTD could not significantly prolong PFS or OS but longer followup suggests a trend towards a late OS advantage(Morgan et al., 2012).

Thalidomide does not compromise successful harvest of stem cells. However, it is associated with an increased risk of venous thromboembolism (VTE) and sensory peripheral neuropathy (PNP). Without thromboprophylaxis, the thrombosis risk is 15-17%, which is more frequent during the first 3 months of treatment and warrants prophylactic anticoagulation. Peripheral neuropathy improves within 3-4 months after dose reduction or cessation of the drug in most patients. However, thalidomide induced PNP may be irreversible if appropriate action is not taken when an emerging neuropathy is encountered. Thalidomide at low dose may be effective in the management of patients with renal failure, but close monitorization for complications is required in patients with serious renal and hepatic failure. Major thalidomide toxicities and the summary of the supportive care guidelines regarding the approach to PNP is given in tables 7 and 8, respectively (Beksac et al., 2008, Bird et al., 2011).

It is clearly understood that thalidomide-based regimens have produced better post-induction response rates (≥PR; 63-82.5%) and PFS than conventional high dose dexamethasone based regimens. However, OS was not prolonged. Poorer response to salvage therapy and decreased survival have been observed in patients who relapsed while on thalidomide. This may be the result of emergence of resistant clones after thalidomide (Rajkumar et al., 2006; Barlogie et al., 2006;, Cavo et al., 2009; Lokhorst et al., 2010; Morgan et al., 2012). Thalidomide-dexamethasone is less active and more toxic than lenalidomide-based regimens and not recommended as first line therapy anymore. However, in countries where lenalidomide is not available as initial first line therapy and in patients with renal failure, thalidomide combinations may be preferred. On the other hand, thalidomide is still being investigated in combination with other drugs as induction and maintenance regimens for both transplant eligible and ineligible MM patients.

Regimen N After induction After ASCT PFS OS Reference
≥PR % ≥VGPR % CR % ≥PR % ≥VGPR % CR %
TD vs. D 103
104
63
41
nr
nr
4
0
nr
nr
nr
nr
nr
nr
nr
nr
nr
nr
Rajkumar 2006
TT2 with T vs.TT2 without T 323
345
nr
nr
nr
nr
nr
nr
nr
nr
nr
nr
62
43
5-year
56%
44%
P=0.01
5-year
65%
65%
P=0.90
Barlogie 2006
TD vs. D 235
235
63
46
43.8
15.8
7.7
2.6
nr
nr
nr
nr
nr
nr
14.9 mos
6.5 mos
P<0.001
nr
nr
Rajkumar 2008
Double ASCT + T vs. Double ASCT 135
135
30
15
68
49
4-year
51%
31%
P=0.001
5-year
69%
53%
P=0.07
Cavo 2009
TAD vs. VAD 268
268
71
57
37
18
3
2
84
76
54
44
14
12
Median
34 mos
22 mos
P<0.001
Median
73 mos
60 mos
P=0.77
Lokhorst 2010
CTD vs.CVAD 555
556
82.5
71
43
27.5
13
8
92
90
62a
74a
50a
37a
Median
27a mos
25a mos
P=0.56
Median
Not reach.
63a mos
P=0.29
Morgan 2012

Table 1.

Phase II- III studies of thalidomide-based regimens as induction therapy before HDT-ASCT

ASCT: autologous stem cell transplantation ; CTD: cyclophosphamide, thalidomide, dexamethasone; CVAD: cyclophosphamide added to VAD (vincristine, doxorubicin, dexamethasone); nr: not reported; TAD: thalidomide, doxorobicin, dexametahsone; TD: thalidomide, dexamethasone; T: thalidomide; TT2: Total therapy-2. a per-protocol analysis


2.2. Bortezomib–based regimens

Bortezomib is an effective inhibitor of proteosome. The ubiquinitin-proteosome pathway plays an important role in intracellular protein homeostasis by regulating degradation of proteins, including mediators of cell cycle progression and apoptosis. Bortezomib also blocks TNF-α mediated upregulation of NF-KB resulting in decreased binding of myeloma cells to bone marrow stromal cells and results in myeloma cell apoptosis. This activity is observed even in cell lines resistant to conventional anti-myeloma therapies. Bortezomib also cleaves DNA repair enzymes increasing the susceptibility of myeloma cells to DNA damaging agents such as alkylating agents and anthracyclines (Hideshima et al., 2001; Cherry et al., 2012).

Bortezomib received FDA approval in 2003 after showing significant activity in relapsed-refractory MM (Jagannath et al., 2004; Richardson et al., 2005). The initial phase II study of single agent bortezomib in previously untreated MM revealed a response rate of 41% (Richardson et al., 2009). Other phase 2 studies incorporating bortezomib ± dexamethasone to induction regimens were consistent with superior responses ranging from 66% to 95% with 6% to 24% CR rates (Jagannath et al., 2005; Harousseau et al., 2006; Rosinol et al., 2007).

The IFM2005-01 Phase III trial compared VD with VAD as induction before ASCT and lenalidomide was given as post-ASCT maintenance in both arms in a randomized fashion. Post-induction at least VGPR (38% vs 15%) and CR/nCR (15% vs 6%) were superior with VD. This response difference was also maintained after ASCT. However, there was only slight improvement in PFS without an OS benefit. Responses with bortezomib were higher regardless of the disease stage or high-risk cytogenetics (Harousseau et al., 2010).

Popat et al. added doxorubicin at escalating doses (0, 4.5 and 9 mg/m2 ) to standard dose bortezomib-dexamethasone (PAD) and demonstrated 95% post-induction response rate with 62% high quality responses (≥ VGPR) (Popat et al., 2008). Addition of pegylated-liposomal doxorubicin to bortezomib-dexamethasone revealed similar responses ( ≥ PR 85% and ≥ VGPR 57.5%) which was further enhanced in patients who underwent ASCT (≥ VGPR 76.6%) (Jakubowiak et al., 2009). A very recent HOVON-65/GMMG-HD4 trial compared pre-transplant PAD (Bortezomib, Adriamycin, Dexamethasone) induction and bortezomib maintenance after ASCT with pre-transplant VAD induction and thalidomide maintenance after ASCT and demonstrated that bortezomib during induction and maintenance significantly improved response rates, quality of response. Similar to other bortezomib containing 3-drug combinations, PFS was significantly improved. Additionally, unlike the other studies, OS was also prolonged in this study (61% vs 55%). Subgroup analysis also indicated that superior outcome with bortezomib was predominantly accomplished in high risk patients presenting with renal failure and del17p (≥ VGPR; 72% vs 43%) (Sonneveld et al., 2012).

Reeder et al. incorporated cyclophosphamide to bortezomib-dexamethasone (CyBorD/VCD) and reported 71% ≥VGPR after 4 cycles and 74% ≥VGPR after ASCT (Reeder et al., 2009). The German Myeloma Study Group reported 84% response rate (≥PR) with 10% CR after 3 cycles of VCD. Over 60% of their patients had high-risk cyctogenetics. The response rate in cytogenetically high-risk patients were 83.7% (del13q) and 90% t(4;14). However, del17p group had lower response rate at 69.2% (Einsele et al., 2009).

These studies demonstrate that bortezomib-based induction studies produce high response rates (≥ PR; 78%-93% and CR 7%-35%) without any adverse effect on stem cell mobilization (Harousseau et al., 2010; Cavo et al.,2010; Rosinol et al., 2012; Sonneveld et al.,2012; Moreau et al., 2010). However, neurotoxicity is a major concern especially when bortezomib is combined with thalidomide. Neurotoxicity can be reduced by reducing bortezomib dose once weekly without affecting the efficacy or by using subcutenous bortezomib (Mateos et al., 2010). Moreover, Moreau et al. used reduced doses of bortezomib (1 mg/m2) and thalidomide (100 mg/d) in vtD regimen and found that this regimen provided higher VGPR rates compared with VD and the dose reduction of both drugs could resulted in reduced incidence of polyneuropathy (Moreau et al., 2011). Major bortezomib toxicities and the summary of the supportive care guidelines regarding the approach to emerging PNP is given in tables 7 and 8, respectively (Bird et al.,2011).

Regimen N After induction After ASCT PFS OS Reference
≥PR % ≥VGPR % CR % ≥PR % ≥VGPR % CR %
VD vs.
VAD
223
218
78.5
63
38
15
15
6
80
77
54
37
35
18
Median
36 mos
30 mos
P=0.06
3-year
81%
77%
P=0.5
Harousseau 2010
IFM 2005-02
VTD vs.
TD
236
238
93
79
62
28
31
11
93
84
82
64
55
41
3-year
68%
56%
P=0.005
3-year
86%
84%
P=0.03
Cavo 2010
GIMEMA-MMY 3006
VBMCP/VBAD+V vs.
VTD vs.
TD
129
130
127
75
85
62
36
60
29
21
35
14
73
77
58
51
65
40
38
46
24
Median
38 mos
27 mos
Not reach.
P=0.006
Nr
Nr
Nr
Rosinol 2012
PETHEMA/
GEM
PAD vs.
VAD
413
414
78
54
42
14
7
2
88
75
62
36
21
9
Median
35 mos
28 mos
P=0.002
5-year
61%
55%
Sonneveld 2012
HOVON65/ GMMG HD4
VD vs.
vtD
99
100
81
88
36
49
31
29
86
89
58
74
22
31
Median
30 mos
26 mos
P=0.22
Nr
nr
Moreau 2011
IFM2007-02

Table 2.

Phase III studies of bortezomib-based regimens in preparation for HDT-ASCT

VD: Bortezomib, dexamethasone; VAD: Vincristine, doxorubicin, dexamethasone; VTD: Bortezomib, thalidomide, dexamethasone; TD: Thalidomide,dexamethasone; VBMCP/VBAD+V: Vincristine, carmustine, melphalan,cyclophosphamide, prednisone/ vincristine, carmustine, doxorubicin, dexamethasone + bortezomib; PAD: Bortezomib, doxorubicin, dexamethasone; vtD: Reduced dose bortezomib, reduced dose thalidomide, dexamethasone; mos: months


Bortezomib has become an important component of therapy for patients with high risk MM associated with del13q and t(4;14) (Richardson et al., 2005; Jagannath et al., 2007). Bortezomib has also proven effective in management of MM patients with renal dysfunction. It has been suggested that in patients with acute renal failure secondary to light chain cast nephropathy VTD can be first choice due to lack of nephrotoxicity. The Mayo Clinic recommends plasma exchange until serum free light chain (FLC)<50 mg/dl and repeated as needed until VTD is fully effective (Rajkumar et al., 2011). Bortezomib is also beneficial for individuals with significant disease related bone-disease due to its inhibitory effect on osteoclastogenesis and stimulatory effect on osteoblast differentiation and proliferation (Zavrski et al., 2005).

Regimen Progression free survival Reference
Overall del13q t(4;14) del(17p)
VD + R ± R vs.
VAD + R ± R
36 mos (median)
30 mos (median)
nr
nr
28mos (median)
16mos (median)
14mos (median)
nr
Avet-Loiseau 2010
PAD + Bort vs.
VAD + T
48% (at 3-yr)
40% (at 3-yr)
40% (at 3-yr)
29% (at 3-yr)
28% (at 3-yr)
20% (at 3-yr)
22% (at 3-yr)
16% (at 3-yr)
Sonneveld 2010
VTD + VTD vs.
TD + TD
68% (at 3-yr)
56% (at 3-yr)
62% (at 3-yr)
46% (at 3-yr)
69% (at 3-yr)
37% (at 3-yr)
nr
nr
Cavo 2010

Table 3.

Impact of bortezomib incorporated into ASCT on PFS according to cytogenetic abnormalities (Cavo 2012)

2.3. Lenalidomide–based regimens

Lenalidomide is another IMID and has more potent in vitro activity; inhibition of angiogenesis, cytokine modulation and T-cell costimulation than thalidomide (Hideshima et al., 2000 ; Haslett et al., 2003). Lenalidomide primarily triggers the caspase-8 mediated apoptotic pathway and also down-regulates NF-KB activity via a mechanism distinct from bortezomib (Mitsiades et al., 2002). Lenalidomide alone (R) or with dexamethasone (RD) has shown significant activity in relapsed/refractory MM. Responses were observed even in patients in whom thalidomide therapy has previously failed (Richardson et al., 2002). Several phase II studies of lenalidomide and dexamethasone +/- chemotherapy have demonstrated response rates ranging 76-91% (Table-4). In a randomized controlled trial lenalidomide plus high dose dexamethasone (RD) (480 mg/28d cycle) was compared with lenalidomide plus low dose dexamethasone (Rd) (160 mg/28d cycle). Patient enrollment to study was not restricted with age or eligibility for ASCT. In each group lenalidomide was administered as 25 mg/d on 1-21 days. In accordance with others, this study demonstrated that lenalidomide in combination with dexamethasone is an efficient initial therapy for MM. Although RD produced higher response rates, this did not result in superior TTP, PFS or OS compared with Rd. The cause of inferior OS with high dose dexamethasone seems to be related to increased deaths due to toxicity, particularly in first 4 months and in eldely patients. The major grade 3 or higher toxicities including thromboembolic events and infections were significantly higher in the high dose dexamethasone group (Rajkumar et al., 2010). The multicenter, placebo controlled SWOG trial has confirmed the superiority of lenalidomide in combination with dexamethasone over dexamethasone alone as initial therapy of MM in terms of response rate and PFS but not in OS. This study received early closure and open-label lenalidomide and dexamethasone was made available to all patients (Zonder et al., 2010). In a retrospective case-control study, RD produced better responses than TD including superior PFS and OS. However, this study was not a randomized trial and the choice of post-induction therapy was not standardized (Gay et al., 2010a). Claritromycin is an antibiotic that has shown efficacy in association with steroids and both thalidomide and lenalidomide. The same investigators added clarithromycin (Bioxin) to Rd(BiRd) and compared with Rd in a case-match study. The have reported significantly better responses with BiRd and the PFS was significantly longer. However, 3- year OS was not statistically different between the two study arms (Gay et al., 2010).

Lenalidomide can cause myelosuppression and concerns have been raised that its use may negatively impact the ability to mobilize stem cells in patients who received lenalidomide as part of their induction therapies (Kumar et al., 2007; Mazumder et al., 2008; Paripati et al., 2008; Popat et al., 2009). It is suggested that stem cells should be collected within 6 months of initiation of lenalidomide therapy and the IMWG recommends that patients >65 years or patients who have received ≥ 4 cycles Rd must undergo stem cell mobilization with cyclophosphamide + G-CSF or G-CSF + plerixafor (Kumar 2007; Kumar 2009). Dose reduction is required in presence of renal impairment. Patients may require thromboprophylaxis due to higher incidence of thromboembolic events. Major lenalidomide toxicities are summarized in table-10 (Bird et al., 2011).

2.4. Novel agent triplet combinations

To enhance response rates and prolong PFS combination of Bortezomib with an IMID has been attempted. Bortezomib-thalidomide-dexamethasone (VTD) resulted better response rates and PFS compared to TD or VD in initial randomized trials (Wang et al., 2005; Cavo et al., 2009). In a Phase III study of VTD compared with TD as induction before and consolidation after double ASCT, VTD was superior to TD in all response categories (CR/n CR; 31% vs 11% ; ≥VGPR; 62% vs 28%) as well as the 3 year estimated PFS (68% vs 56%). Progression free survival was also superior with VTD compared to TD in poor prognostic groups including del13q, increased LDH, age>60 years, t(4;14) ± del17p, increased bone marrow plasma cell ratio and ISS-II and III (Cavo et al., 2010). The results of the PETHEMA/GEM study also provided a strong support to VTD as a highly effective induction regimen compared with a combination chemotherapy containing bortezomib and with TD. Additionally, VTD resulted in a higher post-transplantation CR rate and a significantly longer PFS. However, this did not result in a significant prolongation of OS and could not overcome poor prognosis of high-risk cytogenetics (Rosinol et al., 2012).

Synergy has been demonstrated between bortezomib and lenalidomide. Moreover, both bortezomib and immunomodulatory drugs enhance the activity of dexamethasone. In a phase I study combining these three agents (RVD) in patients with newly diagnosed MM, the maximum tolerated dose was set as lenalidomide 25mg/day, bortezomib 1.3mg/m2, dexamethasone 20mg/day and in phase II portion of the same study, 100% response rate (≥PR) could be obtained with ≥VGPR and CR rates 74% and 37%, respectively. This was the first study to result in 100% response rate. The 18-month PFS and OS were 75% and 97%, respectively (Richardson et al., 2010).

In phase II trials the most promising combinations were either lenalidomide or cyclophosphamide with bortezomib and dexamethasone. A phase II study of four-drug combination VDCR, VDR, VDC and VDC-mod (modification of the cyclophosphamide dose ) was performed to evaluate the feasibility and activity of these combinations. The response seen with VDCR appear to be similar to those seen with VDR or VDC-mod arms (Table-5). However, the toxicities with VDCR appear to be more than the other arms, especially hematological toxicity. This study does not support an advantage of four drug combination (Kumar et al.,2012).

In another phase I/II study RVD was combined with pegylated-doxorubicin (RVDD) and this regimen was highly active and well-tolerated with response rates ≥PR 96% and 95%, ≥VGPR 57% and 65% after 4 and 8 cycles, respectively (Table-6). After a median 15.5 months follow-up, PFS and OS were not reached. The estimated 18-month PFS and OS were 80.8% and 98.6%, respectively. Among patients who proceeded to ASCT and were evaluable for posttransplant response, response rate further improved reaching 85% of patients ≥VGPR and 61% of those with CR/n CR at 3 months after ASCT. In patients who continued RVDD beyond 4 cycles, depth of response further improved reaching 65% ≥VGPR and 35% CR/n CR at the completion of 8 cycles (Jakuboviak et al., 2011).

Regimen N After induction PFS OS Reference
≥PR % ≥VGPR % CR %
RD 34 91 38 6 Rajkumar 2005
RD vs.
Rd
223
222
79
68
42
24
Median
19 mos
25 mos
P=0.026
2-yr
75%
87%
Rajkumar 2010
RD vs.
D
97
95
78
48
3-yr
52%
32%
3-yr
79%
73%
P=0.28
Zonder 2010
RD vs.
TD
228
183
80
61
37.8
15
13.6
3.3
Median
26.7mos
17.1 mos
P=0.036
Median
Notreach
57.2 mos
P=0.018
Gay 2010
BiRd vs.
Rd
72
72
73.6
33
45.8
13.9
Median
48.3 mos
27.5 mos
P=0.044
Median
89.7%
73%
P=0.170
Gay 2010

Table 4.

Results of phase II-III Studies of induction with lenalidomide and dexamethasone

VDCRN=39 VDRN=36 VDCN=31 VDC-modN=16
After 4 cycles
CR
≥VGPR
≥PR
5%
33%
80%
7%
32%
73%
3%
13%
63%
12%
41%
82%
Best response across all cycles (median=6 cycles)
CR
≥VGPR
≥PR
25%
58%
88%
24%
51%
85%
22%
41%
75%
47%
53%
100%
1-yr PFS 86% 83% 93% 100%
After censoring patients going to ASCT
1-yr PFS
1-yr OS
83%
92%
68%
100%
97%
100%
100%
100%
Patients who undergo ASCT
1-yr PFS
1-yr OS
100%
100%
100%
100%
88%
100%
100%
100%

Table 5.

Results of EVOLUTION Study comparing bortezomib-based multi-drug combinations

N Best response after induction (%) Reference
≥PR ≥ VGPR CR
RVD 66 100 67 39 Richardson 2010
VDCR vs.
VDR vs.
VDC vs.
VDC-mod
39
36
31
16
88
85
75
100
58
51
41
53
25
24
22
47
Kumar 2012
RVDD
After 4 cycles
After 8 cycles
74 96
95
57
65
29
35
Jakubowiak 2011

Table 6.

Phase II Trials of triplet or quadruplet lenalidomide-based induction

Thalidomide Lenalidomide Bortezomib
Venous thromboembolism Cytopenias Peripheral neuropathy
Sensory peripheral neuropathy Venous thromboembolism Gastrointestinal toxicity
Constipation Constipation Postural hypotension and pre-syncope secondary to autonomic neuropathy
Hematological toxicity Fatigue Thrombocytopenia
Somnolence Neuropathy Fatigue
Rashes Skin rash Increased incidence of varicella zoster infections
Arrhythmias Muscle cramps
Thyroid dysfunction Thyroid dysfunction
Congenital malformations due to fetal exposure Diarrhea

Table 7.

Major toxicities of novel agents

Grade of neuropathy Bortezomib Thalidomide
Grade 1
Paresthesia, weakness and/or loss of reflexes without painor loss of function
No action No action
Grade 1 with pain or grade 2 interfering with function but not with daily activities Reduce bortezomib dose to 1mg/m2 Reduce thalidomide dose to 50% or suspend thalidomide until disappearence of toxicity, then reinitiate at 50% dose
Grade 2 with pain or grade 3 Suspend bortezomib until disappearence of toxicity then re-initiate at 0.7 mg/m2 and administer once weekly Suspend thalidomide until disappearence of toxicity, then reinitiate at low dose if PNP grade 1
Grade 4 Discontinue Discontinue

Table 8.

Guidelines for the management of bortezomib and thalidomide induced PNP

2.5. Conclusions

High dose melphalan supported by autologous stem cell transplantation after novel agent-based induction regimen is the standard of care for patients younger than 65. The quality of response achieved with induction regimens before ASCT affect PFS and potentially the OS. In this regard, the availability of novel anti-myeloma drugs, thalidomide, bortezomib and lenalidomide has improved the pre-transplantation responses. Recent data suggest that 3-drug induction regimens, containing at least one novel agent result in better responses than 2-drug combinations. The results of studies with combinations of VD with either doxorubicin (PAD), cyclophosphamide (CyBorRd), thalidomide (VTD) or lenalidomide (VRD) have demonstrated that the responses can be further enhanced and PFS ± OS can be improved. Within the 3-drug combinations, bortezomib-dexamethasone combined with thalidomide or cyclophosphamide(VTD or VCD) appear to be the most active regimens. So, 3-6 cycles of a triplet bortezomib based regimen should be considered the standard induction for patients eligible for ASCT. The objective of treatment should be the achievement of a sustained CR with a good quality of life. Current studies concentrate on best approach to combine available drugs to affect long-term disease control as well as consolidation and maintenance after ASCT and minimize the long-term toxicities, especially neurotoxicity. Bortezomib is effective not only in patients with standard risk disease but also in the presence of high risk cytogenetic abnormalities especially in presence of t(4;14). Current question under evaluation is whether to apply or delay ASCT when a CR is achieved with a novel agent induction treatment.

Advertisement

3. Induction therapy for patients ineligible for HDT–ASCT

Melphalan was the first alkylating agent used for treatment of MM and melphalan –prednisone (MP) has been the standard therapy for over 30 years although it yielded only PR in 40-60% of patients with CR <5% and PFS about 18 months and OS 2-3 years. Trials comparing MP with high dose dexamethasone-based combinations revealed no survival advantage (Mateos et al., 2012). During the last decade, with the emergence of novel agents and the studies revealing the importance of achieving VGPR/CR on survival of myeloma patients, the historical goals of induction have changed to achieving a high quality response in elderly patients as well.

3.1. Bendamustine

Bendamustine is a novel bifunctional drug which has similarities to both alkylating agents and purine analogs. It has promising activity in low grade lymphoid malignancies. The East German Study Group conducted a phase III trial comparing bendamustine and prednisone (BP) with standard MP in previously untreated patients with MM who are ineligible for transplantation. Bendamustine –prednisone was superior to MP with respect to CR rate (32% vs 13%, p=0.007) and TTF (14 mos vs 10 mos, p=0.02). There was no significant difference with regard to OS between the two treatment groups and the toxicity profile was comparable (Pönisch et al., 2006). Mainly based on the results of this study, bendamustine is currently approved for treatment of newly diagnosed MM patients who are not candidates for HDT-ASCT and who can not receive thalidomide or bortezomib due to peripheral neuropathy. The same investigators in a recent study demonstrated that bendamustine in combination with bortezomib and prednisone (BPV) is also effective in patients with newly diagnosed MM and renal failure. Eighty-three percent of the patients treated with this protocol responded to therapy and 72% had their renal function improved after treatment (Pönisch et al., 2012).

3.2. Thalidomide–based regimens

Thalidomide incorporated in to the MP regimen (MPT) has been compared with the standard MP regimen in six randomized phase III studies. Each protocol had some minor differences in their schedules which is shown in table-9. The overall response rate (57%-76%) with MPT was significantly higher than MP (31%-48%).The CR rates with MPT ranged between 7%-13%, one study reported ≥ VGPR rate as 27%. In the IFM-I/II studies and in HOVON study, the prolongation in the PFS with MPT was also translated in to OS advantage (Facon et al., 2007; Hulin et al., 2009; Wijermans et al., 2010). Despite in the other three studies the PFS advantage was not translated into OS advantage (Palumbo et al., 2008; Waage et al., 2010; Beksac et al., 2011), a metaanalysis of the pooled data of 1682 patients from these six trials showed that the addition of thalidomide to MP improves OS and PFS in previously untreated elderly patients with multiple myeloma, extending the median survival time by on average 20%. In this metaanalysis, median PFS was prolonged by 5.4 months (HR 0.67 (0.55-0.80) p<0.0001) and the median OS was prolonged 6.6 months (HR 0.82 (0.66-1.02) p=0.004) (Fayers et al., 2011). This improvement was less pronounced in patients aged ≥ 75 years and no favorable effect of thalidomide on OS in this population could be demonstrated. The most frequent grade 3-4 adverse events with MPT protocol were polyneuropathy (6-23%) and VTE (3-12%), infections (10-13%), cardiac complications (2-7%), gastrointestinal events (5%). The discontinuation rate ranged 16-45% (Hulin et al., 2009; Wijermans et al., 2010; Fayers et al., 2011). Based on these results, MPT became one of the new standard therapies for elderly patients with newly diagnosed MM.

Thalidomide-dexamethasone (TD) combination was also compared with MP in 289 elderly patients with MM. Patients achieving stable disease or better were randomly assigned to maintenance therapy with either thalidomide 100 mg daily or interferon alpha-2b. Thalidomide-dexamethasone resulted in a higher proportion of ≥VGPR (26% vs 13%; P=.006) and ORR (68% vs 50%; P=.002) compared with MP. However, PFS was similar (16.7 vs 20.7 months; P=.1) and OS was significantly shorter in the TD group (41.5 vs 49.4 months; P=.024). Decreased survival was more evident in patients older than 75 years due to increased non-disease related deaths during the first year (Ludwig et al., 2009). Combinations with high dose dexamethasone is not recommended for elderly patients especially those ≥ 75 years due to increased toxicity. In a randomized MRC Myeloma IX trial, cyclophosphamide, thalidomide and dexamethasone (CTDa) in which dexamethasone dose was reduced, produced higher response rates than MP but was not associated with improved PFS and OS. Additionally, CTDa was associated with higher rates of adverse events compared to MP (Morgan et al., 2011).

Regimen N Schedule ORR% CR% TTP (months) OS (median) Reference
MPT+T(until PD)
MP
129
126
M: 4mg/m2, d1-7
P: 40mg/m2, d1-7
T: 100mg/d
6 cycles every 4 wks until relapse
76
48
16
2.4
22
15
p=0.004
45
47.6
p=0.79
GIMEMA
Palumbo 2008
MPT
MP
No maintenance
125
196
M:0.25mg/kg, d1-4
P: 2mg/kg, d1-4
T:100-400mg/d
12 cycles every 6 wks
76
35
13
2
28
18
52
33
P=0.0006
IFM-I
Facon 2007
MPT
MP
No maintenance
113
116
M:0.20mg/kg, d1-4
P: 2mg/kg, d1-4
T:100mg/d
12 cycles every 6 wks
62
31
7
1
24
19
P=0.001
44
29
P=0.028
IFM-II
Hulin 2009
MPT+T(until PD)
MP
148
179
M:0.25mg/kg, d1-4
P: 100mg/d, d1-4
T:200-400mg/d
Until plateau every 6 wks
Maintenance T 200mg
57
40
13
4
15
14
ns
29
32
ns
NMSG
Waage 2010
MPT+T(until PD)
MP
165
168
M:0.25mg/kg, d1-5
P: 1mg/kg, d1-5
T:200mg/d
8 cycles every 4 wks
Maintenance T 50mg
66
45
27
10
≥VGPR
13
9
P<0.001
PFS
40
31
P=0.05
HOVON
Wijermans 2010
MPT
MP
No maintenance
62
60
M: 9mg/m2, d1-4
P: 60mg/m2, d1-4
T: 100mg/d
8 cycles every 6 wks
58
38
9
9
21
14
P=0.34
DFS
26
28
P=0.65
TMSG
Beksac 2011

Table 9.

Phase III Studies comparing MPT versus MP for newly diagnosed MM ineligible for HDT-ASCT

M:Melphalan; P:Prednisone; T:Thalidomide; PD:Progressive disease; PFS:Progression free survival; DFS: Disease free survival; GIMEMA: Italian Myeloma Network IFM: Intergroupe Francophone du Mye´lome; NMSG: Nordic Myeloma Study Group; HOVON: Dutch-Belgium Hemato-Oncology Cooperative Group; TMSG:Turkish Myeloma Study Group


3.3. Bortezomib–based regimens

After showing significant efficacy in relapsed-refractory myeloma, bortezomib was also incorporated into trials for initial therapy of MM in transplant ineligible patients. The clinical value of adding bortezomib to the standard MP regimen (VMP) was explored in Velcade as Initial Standard Therapy (VISTA) Study (San Miguel et al., 2008). In this phase III study, 682 newly diagnosed myeloma patients were randomly assigned to receive nine 6-week cycles of melphalan (9 mg/m2) and prednisone (60 mg/m2) on days 1 to 4, either alone or with bortezomib (1.3 mg/m2) on days 1, 4, 8, 11, 22, 25, 29 and 32 during cycles 1 to 4 and on days 1, 8, 22, and 29 during cycles 5 to 9. Addition of bortezomib to MP significantly improved all responses, PFS as well as the OS (Table-10). The main adverse events associated with VMP were neutropenia (40%), thrombocytopenia (37%), peripheral neuropathy (14%), infection (10%) and gastrointestinal events (7%). A recent update of the VISTA study and a subsequent study showed that grade 3-4 hematological and non-hematological adverse events in particular PNP as well as discontinuation of the drug was significantly reduced without affecting the efficacy when once weekly bortezomib schedule was used (Mateos et al., 2010a; Brignhen et al., 2010). In the VISTA trial, patients with high risk cytogenetic profile had similar response rate and OS with the patients with standard risk profile suggesting that addition of bortezomib may overcome the poor prognosis confered to high risk cytogenetics. Another important point of this study was that first-line bortezomib use did not induce more resistant relapse unlike that is seen in thalidomide relapses. The survival data of the VISTA trial was updated at 3 years and 5 years and the survival benefit of VMP protocol remained significant (Mateos et al.,2010a; San Miguel et al., 2011). A part of the VISTA trial investigated the efficacy of VMP protocol in renal impairment excluding patients with Cr >2mg/dl. Response rates with VMP and TTP in both arms did not appear significantly different between patients with GFR ≤ 50 or > 50 mL/min. Moreover, VMP resulted in 44% renal impairment reversal suggesting that this protocol is also an active and well-tolerated treatment option for patients with moderate renal impairment (Dimopoulos et al., 2009).

VMPN=344 MPN=338 p References
ORR (≥PR) 71% 35% <0.001 San Miguel 2008
CR 30% 4% <0.001
TTP 24 mos 16.6 mos <0.001
OS
Median follow-up 16.3 mos
0.008
HRa=0.61
OS
Median follow-up 36.7 mos
Not reached 43 mos <0.001
HRa=0.653
Mateos 2010
OS
Median follow-up 60 mos
56.4 mos 43.1 mos 0.0004
HRa=0.695
San Miguel 2011
3-yr OS 68.5% 54%

Table 10.

Results of the VISTA trial and the long-tem follow-up

a Hazard Ratio


In the PETHEMA trial Mateos et al. have demonstrated that reduced intensity induction with a bortezomib based regimen followed by maintenance is a safe and effective treatment. The investigators compared VMP with VTP (bortezomib, thalidomide, prednisone) as initial therapy in newly diagnosed patients with MM ineligible for transplantation. In both protocols, a reduced intensity bortezomib schedule consisting of one cycle of bortezomib twice per week for 6 weeks followed by five cycles of bortezomib once per week for 5 weeks was used. Patients who completed the six induction cycles were randomly assigned to maintenance therapy with bortezomib plus prednisone (VP) or bortezomib plus thalidomide (VT). The response rates were higher with VTP compared to VMP (CR 28% vs 20% ; ORR 81% vs 80%). Maintenance with VT significantly improved time to progression compared with that for patients who received VP. The support to better response rates with VTP also came from the initial results of UPFRONT study which compared VD, VTD and VMP and revealed better response rates with VTD (Table-11). On the other hand, in both studies patients treated with VTP had more frequent serious adverse events especially PNP and thrombosis (Mateos et al., 2010b; Nievizsky et al., 2011). Combining four drugs (bortezomib, melphalan, prednisone, thalidomide) followed by maintenance with bortezomib-thalidomide (VMPT-VT) was superior to VMP alone in patients with MM who are ineligible for autologous stem-cell transplantation. The 3-years PFS was also improved with VMPT-VT. However, no significant difference in 3-year OS was observed. Additionally, grade 3 to 4 neutropenia, cardiologic events and thromboembolic events were more frequent among patients assigned to the VMPT-VT group than among those assigned to the VMP group (Palumbo et al., 2010). Due to significantly increased adverse events with 4-drug regimen, VMP or MPT are better alternatives for induction of elderly myeloma patients.

N ORR % CR %After induction PFS OS Reference
VMP
VTP
maintenance VP or VT
130
130
80
81
20
28
37 mos
32 mos
60mos median
5-yr 53%
Mateos 2010
VMPT + VT
VMP + no maintenance
254
257
89
81
38
24
P<0.001
3-year
56%
41% p=0.008
3-year
89%
87%
p=0.77
Palumbo 2010
VD
VTD
VMP
168
167
167
73
80
69
24
36
31
NA NA Nievizsky 2011

Table 11.

Phase III Studies comparing bortezomib containing regimens

VD:Bortezomib dexamethasone; VTD: Bortezimib, thalidomide, dexamethasone; VMP: Bortezomib, melphalan, prednisone; VT: Bortezomib, thalidomide; mos: months; NA: Not available


3.4. Lenalidomide–based regimens

In a randomized controlled trial lenalidomide (25 mg/d on d 1-21) plus high dose dexamethasone (480 mg/28d cycle) (RD) produced higher response rates compared with lenalidomide (25 mg/d on d 1-21) plus low dose dexamethasone (Rd) (160 mg/28d cycle). However, this did not translate into superior PFS (Median 19.1 months vs 25.3 months). Moreover, Rd was associated with significantly improved 1-year OS than with RD (96% vs 87%, p=0.0002) and treatment related toxicity was significantly reduced. The cause of inferior OS with high dose dexamethasone seems to be related to increased deaths due to toxicity particularly in first 4 months and in elderly patients. Hence, the advantages of Rd over RD were more pronounced in patients aged > 70 years (Rajkumar et al.,2010; Zonder et al., 2010). A recent double-blind, multicenter, randomized study compared melphalan-prednisone-lenalidomide induction followed by lenalidomide maintenance (MPR-R) with melphalan-prednisone-lenalidomide (MPR) or melphalan-prednisone (MP) followed by placebo. Response rates were superior with MPR-R compared with MPR or MP (ORR, 77% vs 68% vs 50%; CR, 18% vs 13% vs 5%; respectively). MPR-R significantly prolonged PFS in patients with newly diagnosed multiple myeloma who were ineligible for transplantation, with the greatest benefit observed in patients 65 to 75 years of age. This study also underlines the importance of lenalidomide maintenance after MPR induction as another treatment option for elderly myeloma patients The toxicity profile was excessive for frail patients, which negatively affected the efficacy. Main grade 3-4 adverse events of MPR were neutropenia (52-71%), thrombocytopenia (23-38%), infections (10%) and thromboembolism (5%)(Palumbo et al., 2012).

Various circumstances Suggestions
Rapid reversal of spinal cord compression or renal impairment Bortezomib
Pre-existing neuropathy Lenalidomide (MPR, Rd) or bendamustin (BP)
History of venous thromboembolism Bortezomib
In cases with renal failure Thalidomide, bortezomib, bendamustin can be administered at full dose, Lenalidomide requires dose reduction according to creatinin clearence
Contraindications to use of alkyllating agents such as presence myelodysplasia or increased risk of myelosuppression TD or VD can be used instead of MP
Frail patients Prednisone is beter tolerated than dexamethasone

Table 12.

Individualized treatment strategies for non-transplant candidate patients

It is clear that the novel agents have prolonged the survival of patients with MM. However, this benefit is more pronounced in younger patients. Age has been reported to be a negative prognostic factor. It is not because the elderly patients have biologically different disease but because they can not tolerate high intensity therapy protocols, have lower bone marrow reserves, increased tendency for infections and also difficulty in recovering from infections and have more frequent drug toxicities. A patient’s overall physical condition, fraility, comorbidity and disability should be asessed before starting therapy in order to choose the appropriate treatment protocol and dosing. These terms are fully explained in a review by Palumbo et al (Palumbo A et al. 2011). Although the novel agents offer important survival for patients with MM, the incidence of grade 3-4 adverse events and drug discontinuations are significantly higher with combination regimens that are based on novel agents than with traditional chemotherapy regimens. It has been suggested that modifying drug doses at the start of therapy and management of adverse events during the therapy improves tolerability so that the patients can receive the drugs for a longer time to get survival benefit. Secondly, the tolerability of treatment can be further improved with full supportive therapy with bisphosphanates, antivirals, anticoagulants, growth factors and appropriate pain control.

3.5. Conclusions

At present, the induction regimen for patients ineligible for HDT/ASCT is either MP or high/lower dose of Dexamethasone combined with one of the three novel agents (thalidomide or bortezomib or lenalidomide). Selection between these combinations depends on the patients’ presenting symptoms such as presence of neuropathy, renal impairment or the rapidity required to reverse the symptoms. In countries where lenalidomide is not yet allowed as first line therapy, induction can be started with thalidomide or bortezomib containing triple regimens and in case of unresponsiveness or intolerability, lenalidomide can be used as second line therapy. Fraility, comorbidity and disability of the elderly patients should be taken into account before choosing the induction protocol and appropriate dose reductions should be done. Thus, the treatment should be individualized. Melphalan-based regimens are used for a fixed duration (9-18 months) and then observed. However, the duration of treatment with revlimid (Rd) is unclear either continue until relapse or a fixed duration of 18 months has been tested in ongoing phase III trials. Evidence is now emerging that maintenance or continuous therapy with novel agents is improving PFS with a potential to improve OS. However, in elderly patients, it is particularly important to start treatment at a dose that can be tolerated over the long term. Specific recommendations yet can not be made regarding the impact of novel treatment regimens on prognosis of elderly patients with high-risk cytogenetics. Although the Italian study (Palumbo et al., 2010) suggested some PFS benefit in response to VMPT+VT over VMP regarding the high-risk cytogenetics, other studies did not confirm this.

Regimen Usual dosing schedule Reference
Melphalan-Prednisone
(MP-7 day Schedule)
Melphalan 8-10 mg oral days 1-7
Prednisone 60mg/d oral days 1-7
Repeated every 6 weeks
Kyle et al., 2004
Thalidomide-Dexamethasone (Td) Thalidomide 200 mg oral days 1-28
Dexamethasone 40 mg oral days 1,8,15,22
Repeated every 4 weeks
Rajkumar et al., 2006
Lenalidomide-Dexamethasone (Rd) Lenalidomide 25 mg oral days 1-21
Dexamethasone 40 mg oral days 1,8,15,22
Repeated every 4 weeks
Rajkumar et al., 2010
Bortezomib –dexamethasone (Vd) Bortezomib 1.3 mg/m2 iv days 1,8,15,22
Dexamethasone 20 mg on day of and day after bortezomib (or 40 mg days 1,8,15,22)
Repeated every 4 weeks
Harousseau et al., 2006
Melphalan-Prednisone-Thalidomide (MPT) Different MPT protocols are described in table…
Bortezomib-Melphalan-Prednisone (VMP) Bortezomib 1.3 mg/m2 iv days 1,8,15,22
Melphalan 9 mg/ m2 oral days 1-4
Prednisone 60 mg/ m2 oral days 1-4
Repeated every 35 days
San Miguel et al., 2008
Bortezomib-Thalidomide-Dexamethasone (VTD) Bortezomib 1.3 mg/m2 iv days 1,8,15,22
Thalidomide 100-200 mg oral days 1-21
Dexamethasone 20 mg on day of and day after bortezomib (or 40 mg days 1,8,15,22)
Repeated every 4 weeks
Cavo et al., 2009
Cyclophosphamide-Bortezomib-Dexamethasone (CyBorD) Cyclophosphamide 300mg/ m2 oral days 1,8,15,22
Bortezomib 1.3 mg/m2 iv days 1,8,15,22
Dexamethasone 40 mg oral days 1,8,15,22
Repeated every 4 weeks
Reeder et al., 2009
Bortezomib-Cyclophosphamide-Dexamethasone (VCD) Bortezomib 1.3 mg/m2 iv days 1,4,8,11
Cyclophosphamide 900mg/ m2 on day 1 every 3 weeks
Dexamethasone 40 mg on day of and day after bortezomib
Einsele et al., 2009
Bortezomib-Lenalidomide-Dexamethasone (VRD) Bortezomib 1.3 mg/m2 iv days 1,8 and 15
Lenalidomide 25 mg oral days 1-14
Dexamethasone 20 mg on day of and day after bortezomib (or 40 mg days 1,8,15,22)
Repeated every 3 weeks
Richardson et al., 2010
Bortezomib-Melphalan-Prednisone-Thalidomide (VMPT) Bortezomib 1.3 mg/m2 iv days 1,8,15,22 every 35 days
Melphalan 9 mg/ m2 oral days 1-4 every 35 days
Prednisone 60 mg/ m2 oral days 1-4 every 35 days
Thalidomide 50 mg/day
Palumbo et al., 2010
Melphalan-Prednisone-Lenalidomide (MPR-R) Melphalan 0.18 mg/kg on days 1-4 every 4 weeks x 9 cycles
Prednisone 2mg/kg on days 1-4 every 4 weeks x 9 cycles
Lenalidomide 10 mg on days 1-21 then 10mg/d until relapse
Palumbo et al., 2012
Cyclophosphamide-Thalidomide- Dexamethasone (atenuated dose)
(CTD a)
Cyclophosphamide 500mg/week
Thalidomide 50 mg/day for 4 weeks, 50 mg increments every 4 weeks to a maximum 200mg/day
Dexamethasone 20 mg/day on days 1- 4 and 15-18 every 4 weeks
Morgan et al., 2011
Bortezomib-Lenalidomide- Dexamethasone-Cyclophosphamide (VRDC) Bortezomib 1.3 mg/m2 iv days 1,4,8,11
Lenalidomide 15 mg oral days 1-14
Dexamethasone 40 mg days 1,8,15
Cyclophosphamide 500mg/ m2 days 1,8
Kumar et al., 2010

Table 13.

Main treatment protocols in Multiple Myeloma

References

  1. 1. AndersonK. CJagannathSJakubowiakALonialSRajeNAlsinaMGhobrialIKnightREsseltine D & Richardson 2009J Clin Oncol, abstr 8536.
  2. 2. AttalMHarousseauJ. LStoppaA. MSottoJ. JFuzibetJ. GRossiJ. FCasassusPMaisonneuveHFaconTIfrahNPayen C & Bataille R. (1996A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Français du Myélome. N Engl J Med., 11;3352917
  3. 3. Avet-loiseauHLeleuXRousselMMoreauPGuerin-charbonnelCCaillotDMaritGBenboubkerLVoillatLMathiotCKolbBMacroMCampionLWetterwaldMStoppaA. MHulinCFaconTAttalMMinvielle S & Harousseau JL. (2010Bortezomib plus dexamethasone induction improves outcome of patients with t(4;14)myeloma but not outcome of patients with del(17p). J Clin Oncol.,283046304
  4. 4. BarlogieBTricotGAnaissieEShaughnessyJRasmussenEVan RheeFFassasAZangariMHollmigKPineda-romanMLeeCTalamoGThertulienRKiwanEKrishnaSFoxMCrowleyJThalidomide and hematopoietic-cell transplantation for multiple myeloma. N Engl J Med. 2006Mar 9;35410102130
  5. 5. BeksacMDelforge M & Richardson 2008The evolving treatment paradigm of multiple myeloma: From past to present and future. Turk J Hematol., 25(2): 60-70.
  6. 6. BeksacMHaznedarRFiratli-tuglularTOzdoguHAydogduIKonukNSucakGKaygusuzIKarakusSKayaEAliRGulbasZOzetGGoker H & Undar L. (2011Addition of thalidomide to oral melphalan/prednisone in patients with multiple myeloma not eligible for transplantation: results of a randomized trial from the Turkish Myeloma Study Group. Eur J Haematol., 8611622
  7. 7. BirdJ. MOwenR. GDSaSSnowdenJ. APrattGAshcroftJYongKCookGFeylerSDaviesFMorganGCavenaghJLowEBehrensJHaemato-oncologyTask Force of British Committee for Standards in Haematology (BCSH) and UK Myeloma Forum. (2011Guidelines for the diagnosis and management of multiple myeloma 2011. Br J Haematol,15413275
  8. 8. BringhenSLaroccaARossiDCavalliMGenuardiMRiaRGentiliSPatriarcaFNozzoliCLeviAGuglielmelliTBenevoloGCalleaVRizzoVCangialosiCMustoPDe RosaLLiberatiA. MGrassoMFalconeA. PVangelistaACavoMGaidanoGBoccadoro M & Palumbo A. (2010Efficacy and safety of once-weekly bortezomib in multiple myeloma patients. Blood,11623474553
  9. 9. CavoMDi Raimondo F, Zamagni E, Patriarca F, Tacchetti P, Casulli AF, Volpe S, Perrone G, Ledda A, Ceccolini M, Califano C, Bigazzi C, Offidani M, Stefani P,Ballerini F, Fiacchini M, de Vivo A, Brioli A, Tosi P & Baccarani M. (2009Short-term thalidomide incorporated into double autologous stem-cell transplantation improves outcomes in comparison with double autotransplantation for multiple myeloma. J Clin Oncol.,273050017
  10. 10. CavoMRajkumarS. VPalumboAMoreauPOrlowskiRBladéJSezerOLudwigHDimopoulosM. AAttalMSonneveldPBoccadoroMAndersonK. CRichardsonP. GBensingerWJohnsenH. EKroegerNGahrtonGBergsagelP. LVesoleD. HEinseleHJagannathSNiesvizkyRDurieB. GSan Miguel J & Lonial S; International Myeloma Working Group. (2011International Myeloma Working Group consensus approach to the treatment of multiple myeloma patients who are candidates for autologous stem cell transplantation. Blood,11723606373
  11. 11. CavoMTacchettiPPatriarcaFPetrucciM. TPantaniLGalliMDi Raimondo F,Crippa C, Zamagni E, Palumbo A, Offidani M, Corradini P, Narni F, Spadano A,Pescosta N, Deliliers GL, Ledda A, Cellini C, Caravita T, Tosi P & Baccarani M;GIMEMA Italian Myeloma Network. (2010Bortezomib 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,3769758207585
  12. 12. CavoMZamagniETosiPCelliniCCanginiDTacchettiPTestoniNTonelliMDe VivoAPalaretiGTura S & Baccarani M. (2004First-line therapy with thalidomide and dexamethasone in preparation for autologous stem cell transplantation for multiple myeloma. Haematologica, 89782631
  13. 13. CavoMZamagniETosiPTacchettiPCelliniCCanginiDDe VivoATestoniNNicciCTerragnaCGrafoneTPerroneGCeccoliniMTura S & Baccarani M; Bologna 2002 study. (2005Superiority of thalidomide and dexamethasone over vincristine-doxorubicindexamethasone (VAD) as primary therapy in preparation for autologous transplantation for multiple myeloma. Blood ;1061359
  14. 14. Chanan-Khan AA & Giralt S2010Importance of achieving a complete response in multiple myeloma, and the impact of novel agents. J Clin Oncol.,2815261224
  15. 15. CherryB. MKordeNKwokMRoschewski M & Landgren O. (2012Evolving therapeutic paradigms for multiple myeloma: back to the future. Leuk Lymphoma. [Epub ahead of print].
  16. 16. ChildJ. AMorganG. JDaviesF. EOwenR. GBellS. EHawkinsKBrownJDrayson MT & Selby PJ; Medical Research Council Adult Leukaemia Working Party. (2003Highdose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. NEngl J Med, 8;34819187583
  17. 17. DimopoulosM. ARichardsonP. GSchlagRKhuagevaN. KShpilbergOKastritisEKropffMPetrucciM. TDelforgeMAlexeevaJSchotsRMassziTMateosM. VDeraedtWLiuKCakanaAvan de Velde H & San Miguel JF. (2009VMP (Bortezomib, Melphalan, and Prednisone) is active and well tolerated in newly diagnosed patients with multiple myeloma with moderately impaired renal function,and results in reversal of renal impairment: cohort analysis of the phase III VISTA study. J Clin Oncol.,2736608693
  18. 18. EinseleHLiebischPLangerCet alVelcade, intravenous cyclophosphamide and dexamethasone (VCD) induction for previously untreated multiple myeloma (German DSMM XIa Trial). (2009ASH Annual Meeting Abstracts. Abstract 131.
  19. 19. FaconTMaryJ. YHulinCBenboubkerLAttalMPegourieBRenaudMHarousseauJ. LGuillermGChaleteixCDibMVoillatLMaisonneuveHTroncyJDorvauxVMonconduitMMartinCCasassusPJaubertJJardelHDoyenCKolbBAnglaretBGrosboisBYakoub-aghaIMathiot C & Avet-Loiseau H;Intergroupe Francophone du Myélome. (2007Melphalan and prednisone plus thalidomide versus melphalan and prednisone alone or reduced-intensity autologous stem cell transplantation in elderly patients with multiple myeloma (IFM 99-06): a randomised trial. Lancet, 3709594120918
  20. 20. FayersP. MPalumboAHulinCWaageAWijermansPBeksaçMBringhenSMaryJ. YGimsingPTermorshuizenFHaznedarRCaravitaTMoreauPTuressonIMustoPBenboubkerLSchaafsmaMSonneveld P & Facon T; Nordic Myeloma Study Group; Italian Multiple Myeloma Network; Turkish Myeloma Study Group; Hemato-Oncologie voor Volwassenen Nederland; Intergroupe Francophone du Myélome; European Myeloma Network. (2011Thalidomide for previously untreated elderly patients with multiple myeloma: meta-analysis of 1685 individual patient data from 6 randomized clinical trials. Blood, 1185123947
  21. 21. FermandJ. PKatsahianSDivineMLeblondVDreyfusFMacroMArnulfBRoyerBMarietteXPertuisetEBelangerCJanvierMChevretSBrouet JC & Ravaud P; Group Myelome-Autogreffe. (2005High-dose therapy and autologous blood stemcell transplantation compared with conventional treatment in myeloma patients aged 55 to 65 years: long-term results of a randomized control trial from the Group Myelome-Autogreffe. J Clin Oncol, 20;2336922733
  22. 22. GayFHaymanS. RLacyM. QBuadiFGertzM. AKumarSDispenzieriAMikhaelJ. RBergsagelP. LDingliDReederC. BLustJ. ARussellS. JRoyVZeldenrustS. RWitzigT. EFonsecaRKyleR. AGreippP. RStewart AK & Rajkumar SV. (2010Lenalidomide plus dexamethasone versus thalidomide plus dexamethasone in newly diagnosed multiple myeloma: a comparative analysis of 411 patients. Blood,1157134350
  23. 23. GayFRajkumarS. VColemanMKumarSMarkTDispenzieriAPearseRGertzM. ALeonardJLacyM. QChen-kiangSRoyVJayabalanD. SLustJ. AWitzigT. EFonsecaRKyleR. AGreippP. RStewart AK & Niesvizky R. (2010Clarithromycin (Biaxin)-lenalidomide-low-dose dexamethasone (BiRd) versus lenalidomide-low-dose dexamethasone (Rd) for newly diagnosed myeloma. Am J Hematol.,8596649
  24. 24. HarousseauJ. LAttal M & Avet-Loiseau H. (2009The role of complete response in multiple myeloma. Blood, 11415313946
  25. 25. HarousseauJ. LAttalMAvet-loiseauHMaritGCaillotDMohtyMLenainPHulinCFaconTCasassusPMichalletMMaisonneuveHBenboubkerLMaloiselFPetillonM. OWebbIMathiot C & Moreau 2010Bortezomib 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.,28(30):4621-9.
  26. 26. HarousseauJ. LAttalMAvet-loiseauHMaritGCaillotDMohtyMLenainPHulinCFaconTCasassusPMichalletMMaisonneuveHBenboubkerLMaloiselFPetillonM. OWebbIMathiot C & Moreau 2010Bortezomib 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.,28(30):4621-9.
  27. 27. HarousseauJ. LAttalMLeleuXTroncyJPegourieBStoppaA. MHulinCBenboubkerLFuzibetJ. GRenaudMMoreau P & Avet-Loiseau H. (2006Bortezomib 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,91111498505
  28. 28. HaslettP. AHanekomW. AMuller G & Kaplan G. (2003Thalidomide and a thalidomide analogue drug costimulate virus-specific CD8+ T cells in vitro. J Infect Dis.,187694655
  29. 29. HideshimaTChauhanDShimaYRajeNDaviesF. ETaiY. TTreonS. PLinBSchlossmanR. LRichardsonPMullerGStirling DI & Anderson KC. (2000Thalidomide and its analogs overcome drug resistance of human multiple myeloma cells to conventional therapy. Blood,969294350
  30. 30. HideshimaTChauhanDShimaYRajeNDaviesF. ETaiY. TTreonS. PLinBSchlossmanR. LRichardsonPMullerGStirling DI & Anderson KC. (2000Thalidomide and its analogs overcome drug resistance of human multiple myeloma cells to conventional therapy. Blood,969294350
  31. 31. HideshimaTRichardsonPChauhanDPalombellaV. JElliottP. JAdams J &Anderson KC. (2001The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res.,61730716
  32. 32. HulinCFaconTRodonPPegourieBBenboubkerLDoyenCDibMGuillermGSallesBEschardJ. PLenainPCasassusPAzaïsIDecauxOGarderetLMathiotCFontanJLafonIVirion JM & Moreau 2009Efficacy of melphalan and prednisone plus thalidomide in patients older than 75 years with newly diagnosed multiple myeloma: IFM 01/01 trial. J Clin Oncol., 27(22):3664-70.
  33. 33. JagannathSBarlogieBBerensonJSiegelDIrwinDRichardsonP. GNiesvizkyRAlexanianRLimentaniS. AAlsinaMAdamsJKauffmanMEsseltineD. LSchenkein DP & Anderson KC. (2004A phase 2 study of two doses of bortezomib in relapsed or refractory myeloma. Br J Haematol.,127216572
  34. 34. JagannathSDurieB. GWolfJCamachoEIrwinDLutzkyJMckinleyMGabayanEMazumderASchenkein D&Crowley J. (2005Bortezomib therapy alone and in combination with dexamethasone for previously untreated symptomatic multiple myeloma. Br J Haematol.,129677683
  35. 35. JagannathSRichardsonP. GSonneveldPSchusterM. WIrwinDStadtmauerE. AFaconTHarousseauJ. LCowan JM & Anderson KC. (2007Bortezomib appears to overcome the poor prognosis conferred by chromosome 13 deletion in phase 2 and 3 trials. Leukemia,2111517
  36. 36. JakubowiakA. JGriffithK. AReeceD. EHofmeisterC. CLonialSZimmermanT. MCampagnaroE. LSchlossmanR. LLaubachJ. PRajeN. SAndersonTMietzelM. AHarveyC. KWearS. MBarrickmanJ. CTendlerC. LEsseltineD. LKelleyS. LKaminskiM. SAnderson KC & Richardson PG. (2011Lenalidomide, bortezomib, pegylated liposomal doxorubicin, and dexamethasone in newly diagnosed multiple myeloma: a phase 1/2 Multiple Myeloma Research Consortium trial. Blood,118353543
  37. 37. JakubowiakA. JKendallTAl-zoubiAKhaledYMineishiSAhmedACampagnaroEBrozoCBraunTTalpaz M & Kaminski MS. (2009Phase II trial of combination therapy with bortezomib, pegylated liposomal doxorubicin, and dexamethasone in patients with newly diagnosed myeloma. J Clin Oncol,2730501522
  38. 38. KastritisEZervasKSymeonidisATerposEDelimbassiSAnagnostopoulosNMichaliEZomasAKatodritouEGikaDPouliAChristoulasDRoussouMKartasisZEconomopoulos T & Dimopoulos MA. (2009Improved survival of patients with multiple myeloma after the introduction of novel agents and the applicability of the International Staging System (ISS): an analysis of the Greek Myeloma Study Group (GMSG). Leukemia, 23611527
  39. 39. KorethJCutlerC. SDjulbegovicBBehlRSchlossmanR. LMunshiN. CRichardsonP. GAndersonK. CSoiffer RJ & Alyea EP 3rd. (2007High-dose therapy with single autologous transplantation versus chemotherapy for newly diagnosed multiple myeloma: A systematic review and meta-analysis of randomized controlled trials. Biol Blood Marrow Transplant, 13218396
  40. 40. KumarSDispenzieriALacyM. QHaymanS. RBuadiF. KGastineauD. ALitzowM. RFonsecaRRoyVRajkumar SV& Gertz MA. (2007Impact of lenalidomide therapy on stem cell mobilization and engraftment post-peripheral blood stem cell transplantation in patients with newly diagnosed myeloma. Leukemia, 21203542
  41. 41. KumarSFlinnIRichardsonP. GHariPCallanderNNogaS. JStewartA. KTurturroFRifkinRWolfJEstevamJMulliganGShiHWebb IJ & Rajkumar SV. (2012Randomized, multicenter, phase 2 study (EVOLUTION) of combinations of bortezomib,dexamethasone, cyclophosphamide, and lenalidomide in previously untreated multiple myeloma. Blood,11919437582
  42. 42. KumarSGiraltSStadtmauerE. AHarousseauJ. LPalumboABensingerWComenzoR. LLentzschSMunshiNNiesvizkyRSan Miguel J, Ludwig H, Bergsagel L, Blade J,Lonial S, Anderson KC, Tosi P, Sonneveld P, Sezer O, Vesole D, Cavo M, Einsele H,Richardson PG, Durie BG & Rajkumar SV; International Myeloma Working Group. (2009Mobilization in myeloma revisited: IMWG consensus perspectives on stem cell collection following initial therapy with thalidomide-, lenalidomide-, or bortezomib-containing regimens. Blood, 27;1149172935
  43. 43. KumarS. KRajkumarS. VDispenzieriALacyM. QHaymanS. RBuadiF. KZeldenrustS. RDingliDRussellS. JLustJ. AGreippP. RKyle RA & Gertz MA. (2008Improved survival in multiple myeloma and the impact of novel therapies. Blood,1115251620
  44. 44. Kyle RA & Rajkumar SV2004Multiple myeloma. N Engl J Med.,35118186073
  45. 45. LokhorstH. MSchmidt-wolfISonneveldPVan Der HoltBMartinHBargeRBertschUSchlenzkaJBosG. MCroockewitSZweegmanSBreitkreutzIJoostenPScheidCVan Marwijk-kooyMSalwenderH. JVan OersM. HSchaafsmaRNaumannRSinnigeHBlauIDelforgeMDe WeerdtOWijermansPWittebolSDuersenUVellenga E & Goldschmidt H; Dutch-Belgian HOVON; German GMMG. (2008Thalidomide in induction treatment increases the very good partial response rate before and after high-dose therapy in previously untreated multiple myeloma. Haematologica,9311247
  46. 46. LokhorstH. MVan Der HoltBZweegmanSVellengaECroockewitSVan OersM. Hvon dem Borne P, Wijermans P, Schaafsma R, de Weerdt O, Wittebol S, Delforge M, Berenschot H, Bos GM, Jie KS, Sinnige H, van Marwijk-Kooy M, Joosten P, Minnema MC, van Ammerlaan R & Sonneveld P; Dutch-Belgian Hemato-Oncology Group (HOVON). (2010A randomized phase 3 study on the effect of thalidomide combined with adriamycin,dexamethasone, and high-dose melphalan, followed by thalidomide maintenance in patients with multiple myeloma. Blood, 1156111320
  47. 47. LudwigHBeksacMBladéJCavenaghJCavoMDelforgeMDimopoulosMDrachJEinseleHFaconTGoldschmidtHHarousseauJ. LHessUKropffMLeal da Costa F, Louw V, Magen-Nativ H, Mendeleeva L, Nahi H, Plesner T, San-Miguel J,Sonneveld P, Udvardy M, Sondergeld P & Palumbo A. (2011Multiple myeloma treatment strategies with novel agents in 2011: a European perspective. Oncologist,164388403
  48. 48. LudwigHHajekRTóthováEDrachJAdamZLabarBEgyedMSpickaIGisslingerHGreilRKuhnIZojer N & Hinke A. (2009Thalidomide-dexamethasone compared with melphalan-prednisolone in elderly patients with multiple myeloma. Blood, 11315343542
  49. 49. Mateos MV & San Miguel JFOld and new treatments in non-transplant candidate newly diagnosed MM patients. Hematology education programme fort he annual congress of the European Hematology Association. (2012Haematologica,61221227
  50. 50. MateosM. VOriolAMartínez-lópezJGutiérrezNTeruelA. IDe PazRGarcía-larañaJBengoecheaEMartínAMediavillaJ. DPalomeraLDe ArribaFGonzálezYHernándezJ. MSuredaABelloJ. LBargayJPeñalverF. JRiberaJ. MMartín-mateosM. LGarcía-sanzRCibeiraM. TRamosM. LVidrialesM. BPaivaBMontalbánM. ALahuertaJ. JBladé J & Miguel JF.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.,111093441
  51. 51. MateosM. VOriolAMartínez-lópezJGutiérrezNTeruelA. IDe PazRGarcía-larañaJBengoecheaEMartínAMediavillaJ. DPalomeraLDe ArribaFGonzálezYHernándezJ. MSuredaABelloJ. LBargayJPeñalverF. JRiberaJ. MMartín-mateosM. LGarcía-sanzRCibeiraM. TRamosM. LVidrialesM. BPaivaBMontalbánM. ALahuertaJ. JBladé J & Miguel JF. (2010Bortezomib, 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.,111093441
  52. 52. MateosM. VRichardsonP. GSchlagRKhuagevaN. KDimopoulosM. AShpilbergOKropffMSpickaIPetrucciM. TPalumboASamoilovaO. SDmoszynskaAAbdulkadyrovK. MSchotsRJiangBEsseltineD. LLiuKCakanaAvan de Velde H & San Miguel JF. (2010Bortezomib 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.,2813225966
  53. 53. MazumderAKaufmanJNiesvizkyRLonialSVesole D & Jagannath S. (2008Effect of lenalidomide therapy on mobilization of peripheral blood stem cells in previously untreated multiple myeloma patients. Leukemia, 22612801
  54. 54. MitsiadesNMitsiadesC. SPoulakiVChauhanDRichardsonP. GHideshimaTMunshiN. CTreon SP & Anderson KC. (2002Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: therapeutic implications. Blood,9912452530
  55. 55. MoreauPAvet-loiseauHFaconTAttalMTiabMHulinCDoyenCGarderetLRandriamalalaEAraujoCLepeuGMaritGCaillotDEscoffreMLioureBBenboubkerLPégouriéBKolbBStoppaA. MFuzibetJ. GDecauxODibMBerthouCChaleteixCSebbanCTraulléCFontanJWetterwaldMLenainPMathiot C & Harousseau JL. (2011Bortezomib plus dexamethasone versus reduced-dose bortezomib, thalidomide plus dexamethasone as induction treatment before autologous stem cell transplantation in newly diagnosed multiple myeloma. Blood,1182257528
  56. 56. MoreauPHulinCMaritGCaillotDFaconTLenainPBerthouCPégouriéBStoppaA. MCasassusPMichalletMBenboubkerLMaisonneuveHDoyenCLeyvrazSMathiotCAvet-loiseauHAttal M & Harousseau JL; IFM group. (2010Stem cell collection in patients with de novo multiple myeloma treated with thecombination of bortezomib and dexamethasone before autologous stem cell transplantation according to IFM 2005-01 trial. Leukemia,24612335
  57. 57. MorganG. JDaviesF. EGregoryW. MBellS. ESzubertA. JNavarro Coy N, Cook G, Feyler S, Johnson PR, Rudin C, Drayson MT, Owen RG, Ross FM, Russell NH, Jackson GH & Child JA; National Cancer Research Institute Haematological Oncology Clinical Studies Group. (2012Cyclophosphamide, thalidomide, and dexamethasone as induction therapy for newly diagnosed multiple myeloma patients destined for autologous stem-cell transplantation: MRC Myeloma IX randomized trial results. Haematologica,97344250
  58. 58. MorganG. JDaviesF. EGregoryW. MRussellN. HBellS. ESzubertA. JNavarro Coy N, Cook G, Feyler S, Byrne JL, Roddie H, Rudin C, Drayson MT, Owen RG, Ross FM, Jackson GH & Child JA; NCRI Haematological Oncology Study Group. Cyclophosphamide, thalidomide, and dexamethasone (CTD) as initial therapy for patients with multiple myeloma unsuitable for autologous transplantation. Blood, 118512318
  59. 59. NiesvizkyRFlinnI. WRifkinR. MGabrailN. YCharuVClowneyBEssellJGaffarY. AWarrT. ANeuwirthRCorzo D & Reeves JA. (2010Phase 3b UPFRONT Study: Safety and Efficacy of Weekly Bortezomib Maintenance Therapy After Bortezomib-Based Induction Regimens In Elderly, Newly Diagnosed Multiple Myeloma Patients. Blood (ASH Annual Meeting Abstracts) 116: Abstract 619.
  60. 60. Palumbo A & Anderson K2011Multiple myeloma. N Engl J Med.,36411104660
  61. 61. PalumboABringhenSCaravitaTMerlaECapparellaVCalleaVCangialosiCGrassoMRossiniFGalliMCatalanoLZamagniEPetrucciM. TDe StefanoVCeccarelliMAmbrosiniM. TAvontoIFalcoPCicconeGLiberatiA. MMusto P & Boccadoro M; Italian Multiple Myeloma Network, GIMEMA. (2006Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: randomised controlled trial. Lancet, 367951382531
  62. 62. PalumboABringhenSLiberatiA. MCaravitaTFalconeACalleaVMontanaroMRiaRCapaldiAZambelloRBenevoloGDerudasDDoreFCavalloFGayFFalcoPCicconeGMustoPCavo M & Boccadoro M. (2008Oral melphalan, prednisone, and thalidomide in elderly patients with multiple myeloma: updated results of a randomized controlled trial. Blood,1128310714
  63. 63. PalumboABringhenSLudwigHDimopoulosM. ABladéJMateosM. VRosiñolLBoccadoroMCavoMLokhorstHZweegmanSTerposEDaviesFDriessenCGimsingPGramatzkiMHàjekRJohnsenH. ELeal Da Costa F, Sezer O, Spencer A,Beksac M, Morgan G, Einsele H, San Miguel JF & Sonneveld 2011Personalized therapy in multiple myeloma according to patient age and vulnerability: a report of the European Myeloma Network (EMN). Blood,118(17):4519-29.
  64. 64. PalumboABringhenSRossiDCavalliMLaroccaARiaROffidaniMPatriarcaFNozzoliCGuglielmelliTBenevoloGCalleaVBaldiniLMorabitoFGrassoMLeonardiGRizzoMFalconeA. PGottardiDMontefuscoVMustoPPetrucciM. TCiccone G & Boccadoro M. (2010Bortezomib-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.,283451019
  65. 65. PalumboAGiacconeLBertolaAPregnoPBringhenSRusCTrioloSGalloEPileri A & Boccadoro M. (2001Low-dose thalidomide plus dexamethasone is an effective salvage therapy for advanced myeloma. Haematologica.,864399403
  66. 66. PalumboAHajekRDelforgeMKropffMPetrucciM. TCatalanoJGisslingerHWiktor-jedrzejczakWZodelavaMWeiselKCascavillaNIosavaGCavoMKloczkoJBladéJBeksacMSpickaIPlesnerTRadkeJLangerCBen Yehuda D, Corso A, Herbein L, Yu Z, Mei J, Jacques C & Dimopoulos MA; MM-015 Investigators. (2012Continuous lenalidomide treatment for newly diagnosed multiple myeloma. N Engl J Med., 36619175969
  67. 67. ParipatiHStewartA. KCabouSDueckAZepedaV. JPiroozNEhlenbeckCReederCSlackJLeisJ. FBoesigerJTorloniA. SFonseca R & Bergsagel PL. (2008Compromised stem cell mobilization following induction therapy with lenalidomide in myeloma. Leukemia, 22612824
  68. 68. PönischWAndreaMWagnerIHammerschmidtDKreibichUSchwarzerAZehrfeldTSchwarzMWinkelmannCPetrosSBachmannALindner T & Niederwieser D. (2012Successful treatment of patients with newly diagnosed/untreated multiple myeloma and advanced renal failure using bortezomib in combination with bendamustine and prednisone. J Cancer Res Clin Oncol.,1388140512
  69. 69. PönischWMitrouP. SMerkleKHeroldMAssmannMWilhelmGDachseltKRichterPSchirmerVSchulzeASubertRHarkselBGrobeNStelzerESchulzeMBittrichAFreundMPasoldRFriedrichTHelbig W & Niederwieser D; East German Study Group of Hematology and Oncology (OSHO). (2006Treatment of bendamustine and prednisone in patients with newly diagnosed multiple myeloma results in superior complete response rate, prolonged time totreatment failure and improved quality of life compared to treatment with melphalan and prednisone--a randomized phase III study of the East German Study Group of Hematology and Oncology (OSHO). J Cancer Res Clin Oncol.,132420512
  70. 70. PopatROakerveeH. EHallamSCurryNOdehLFootNEsseltineD. LDrakeMMorris C & Cavenagh JD. (2008Bortezomib, doxorubicin and dexamethasone (PAD) front-line treatment of multiple myeloma: updated results after long-term follow-up. Br J Haematol,14145126
  71. 71. PopatUSalibaRThandiRHosingCQazilbashMAnderliniPShpallEMcmannisJKörblingMAlousiAAnderssonBNietoYKebriaeiPKhouriIDe LimaMWeberDThomasSWangMJonesRChamplin R & Giralt S. (2009Impairment of filgrastim-induced stem cell mobilization after prior lenalidomide in patients with multiple myeloma. Biol Blood Marrow Transplant, 1571823
  72. 72. RajkumarS. VBloodEVesoleDFonseca R & Greipp PR; Eastern Cooperative Oncology Group. (2006Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group. J Clin Oncol.,2434316
  73. 73. RajkumarS. VHaymanSGertzM. ADispenzieriALacyM. QGreippP. RGeyerSIturriaNFonsecaRLustJ. AKyle RA & Witzig TE. (2002Combination therapy with thalidomide plus dexamethasone for newly diagnosed myeloma. J Clin Oncol.,2021431923
  74. 74. RajkumarS. VHaymanS. RLacyM. QDispenzieriAGeyerS. MKabatBZeldenrustS. RKumarSGreippP. RFonsecaRLustJ. ARussellS. JKyleR. AWitzig TE & Gertz MA. (2005Combination therapy with lenalidomide plus dexamethasone (Rev/Dex) for newly diagnosed myeloma. Blood,1061340503
  75. 75. RajkumarS. VJacobusSCallanderN. SFonsecaRVesoleD. HWilliamsM. EAbonourRSiegelD. SKatz M & Greipp PR; Eastern Cooperative Oncology Group. (2010Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial. Lancet Oncol.,1112937
  76. 76. RajkumarS. VRosiñolLHusseinMCatalanoJJedrzejczakWLucyLOlesnyckyjMYuZKnightRZeldis JB & Bladé J. (2008Multicenter, randomized, double-blind, placebo-controlled study of thalidomide plus dexamethasone compared with dexamethasone as initial therapy for newly diagnosed multiple myeloma. J Clin Oncol.,261321717
  77. 77. RajkumarS. V2012Multiple myeloma: 2012 update on diagnosis, risk-stratification, and management. Am J Hematol., 8717888
  78. 78. ReederC. BReeceD. EKukretiVChenCTrudelSHentzJNobleBPiroozN. ASpongJ. EPizaJ. GZepedaV. HMikhaelJ. RLeisJ. FBergsagelP. LFonseca R & Stewart AK. (2009Cyclophosphamide, bortezomib and dexamethasone induction for newly diagnosed multiple myeloma: high response rates in a phase II clinical trial. Leukemia,237133741
  79. 79. RichardsonP. GBarlogieBBerensonJSinghalSJagannathSIrwinDRajkumarS. VHideshimaTXiaoHEsseltineDSchenkein D & Anderson KC; SUMMIT Investigators. (2005Clinical factors predictive of outcome with bortezomib in patients with relapsed,refractory multiple myeloma. Blood,1069297781
  80. 80. RichardsonP. GSchlossmanR. LWellerEHideshimaTMitsiadesCDaviesFLeBlanc R, Catley LP, Doss D, Kelly K, McKenney M, Mechlowicz J, Freeman A,Deocampo R, Rich R, Ryoo JJ, Chauhan D, Balinski K, Zeldis J & Anderson KC. (2002Immunomodulatory drug CC-5013 overcomes drug resistance and is well tolerated in patients with relapsed multiple myeloma. Blood,100930637
  81. 81. RichardsonP. GSonneveldPSchusterM. WIrwinDStadtmauerE. AFaconTHarousseauJ. LBen-yehudaDLonialSGoldschmidtHReeceDSan-miguelJ. FBladéJBoccadoroMCavenaghJDaltonW. SBoralA. LEsseltineD. LPorterJ. BSchenkein D & Anderson KC; Assessment of Proteasome Inhibition for Extending Remissions (APEX) Investigators. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med.,352248724248798
  82. 82. RichardsonP. GWellerELonialSJakubowiakA. JJagannathSRajeN. SAviganD. EXieWGhobrialI. MSchlossmanR. LMazumderAMunshiN. CVesoleD. HJoyceRKaufmanJ. LDossDWarrenD. LLundeL. EKasterSDelaneyCHideshimaTMitsiadesC. SKnightREsseltine DL & Anderson KC. (2010Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma. Blood,116567986
  83. 83. RichardsonP. GXieWMitsiadesCChanan-khanA. ALonialSHassounHAviganD. EOaklanderA. LKuterD. JWenP. YKesariSBriembergH. RSchlossmanR. LMunshiN. CHeffnerL. TDossDEsseltineD. LWellerEAnderson KC & Amato AA. (2009Single-agent bortezomib in previously untreated multiple myeloma: efficacy,characterization of peripheral neuropathy, and molecular correlations with response and neuropathy.J Clin Oncol.,2721351825
  84. 84. RosiñolLOriolAMateosM. VSuredaAGarcía-sánchezPGutiérrezNAlegreALahuertaJ. JDe La RubiaJHerreroCLiuXVan de Velde H, San Miguel J & Bladé J. (2007Phase II PETHEMA trial of alternating bortezomib and dexamethasone as induction regimen before autologous stem-cell transplantation in younger patients with multiple myeloma: efficacy and clinical implications of tumor response kinetics. J Clin Oncol.,252844528
  85. 85. RosiñolLOriolATeruelA. IHernándezDLópez-jiménezJDe La RubiaJGranellMBesalduchJPalomeraLGonzálezYEtxebesteM. ADíaz-mediavillaJHernándezM. TDe ArribaFGutiérrezN. CMartín-ramosM. LCibeiraM. TMateosM. VMartínezJAlegreALahuertaJ. JSan Miguel J & Bladé J; on behalf of the Programa para el Estudio y la Terapéutica de las Hemopatías Malignas/Grupo Español de Mieloma (PETHEMA/GEM) group. (2012Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study. Blood,120815891596
  86. 86. San Miguel JFSchlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M,Spicka I, Petrucci MT, Palumbo A, Samoilova OS, Dmoszynska A, Abdulkadyrov KM, Schots R, Jiang B, Mateos MV, Anderson KC, Esseltine DL, Liu K, Cakana A, van de Velde H & Richardson PG; VISTA Trial Investigators. (2008Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med, 359990617
  87. 87. SinghalSMehtaJDesikanRAyersDRobersonPEddlemonPMunshiNAnaissieEWilsonCDhodapkarMZeddis J& Barlogie B. (1999Antitumor activity of thalidomide in refractory multiple myeloma.N Engl J Med., 34121156571
  88. 88. SonneveldPSchmidt-wolfI. GVan Der HoltBEl JarariLBertschUSalwenderHZweegmanSVellengaEBroylABlauI. WWeiselK. CWittebolSBosG. MStevens-kroefMScheidCPfreundschuhMHoseDJauchAVan Der VeldeHRaymakersRSchaafsmaM. RKerstenM. JVan Marwijk-kooyMDuehrsenULindemannWWijermansP. WLokhorst HM & Goldschmidt HM. (2012Bortezomib Induction and Maintenance Treatment in Patients With Newly Diagnosed Multiple Myeloma: Results of the Randomized Phase III HOVON-65/ GMMG-HD4 Trial. J Clin Oncol.,3024294655
  89. 89. SonneveldPSchmidt-wolfI. GVan Der HoltBEl JarariLBertschUSalwenderHZweegmanSVellengaEBroylABlauI. WWeiselK. CWittebolSBosG. MStevens-kroefMScheidCPfreundschuhMHoseDJauchAVan Der VeldeHRaymakersRSchaafsmaM. RKerstenM. JVan Marwijk-kooyMDuehrsenULindemannWWijermansP. WLokhorst HM& Goldschmidt HM. (2012Bortezomib Induction and Maintenance Treatment in Patients With Newly Diagnosed Multiple Myeloma: Results of the Randomized Phase III HOVON-65/ GMMG-HD4 Trial. J Clin Oncol. 2012 Aug 20;3024294655
  90. 90. WaageAGimsingPFayersPAbildgaardNAhlbergLBjörkstrandBCarlsonKDahlI. MForsbergKGulbrandsenNHaukåsEHjertnerOHjorthMKarlssonTKnudsenL. MNielsenJ. LLinderOMellqvistU. HNesthusIRolkeJStrandbergMSørbøJ. HWisløffFJuliusson G & Turesson I; Nordic Myeloma Study Group. (2010Melphalan and prednisone plus thalidomide or placebo in elderly patients with multiple myeloma. Blood, 1169140512
  91. 91. WangMGiraltSDelasalleKHandy B & Alexanian R. (2007Bortezomib in combination with thalidomide-dexamethasone for previously untreated multiple myeloma. Hematology, 1232359
  92. 92. WijermansPSchaafsmaMTermorshuizenFAmmerlaanRWittebolSSinnigeHZweegmanSvan Marwijk Kooy M, van der Griend R, Lokhorst H & Sonneveld P; Dutch-Belgium Cooperative Group HOVON. (2010Phase III study of the value of thalidomide added to melphalan plus prednisone in elderly patients with newly diagnosed multiple myeloma: the HOVON 49 Study. J Clin Oncol.,281931606
  93. 93. ZavrskiIKrebbelHWildemannBHeiderUKaiserMPossinger K & Sezer O. (2005Proteasome inhibitors abrogate osteoclast differentiation and osteoclast function. Biochem Biophys Res Commun.,33312005
  94. 94. ZonderJ. ACrowleyJHusseinM. ABolejackVMoore DF Sr, Whittenberger BF,Abidi MH, Durie BG & Barlogie B. (2010Lenalidomide and high-dose dexamethasone compared with dexamethasone as initial therapy for multiple myeloma: a randomized Southwest Oncology Group trial (S0232). Blood,11626583841

Written By

Sule Mine Bakanay and Meral Beksac

Submitted: 09 October 2012 Published: 10 April 2013