Thalidomide, bortezomib, and lenalidomide have recently changed the treatment paradigm of myeloma. In young, newly diagnosed patients, the combination of thalidomide and dexamethasone has been widely used as induction treatment before autologous stem cell transplantation (ASCT). In 2 randomized studies, consolidation or maintenance with low-dose thalidomide has extended both progression-free and overall survival in patients who underwent ASCT at diagnosis. In elderly, newly diagnosed patients, 3 independent randomized studies have reported that the oral combination of melphalan and prednisone plus thalidomide (MPT) is better than the standard melphalan and prednisone (MP). These studies have shown better progression-free survival, and 2 have shown improved overall survival for patients assigned to MPT. In refractory-relapsed disease, combinations including thalidomide with dexamethasone, melphalan, doxorubicin, or cyclophosphamide have been extensively investigated. The risks of side effects are greater when thalidomide is used in combination with other drugs. Thromboembolism and peripheral neuropathy are the major concern. The introduction of anticoagulant prophylaxis has reduced the rate of thromboembolism to less than 10%. Immediate thalidomide dose reduction or discontinuation when paresthesia is complicated by pain or motor deficit has decreased the severity of neuropathy. Future studies will define the most effective or the best sequence of combinations which could improve life expectancy.

Thalidomide was initially used in Europe as a sedative, tranquilizer, and antiemetic for emesis gravidarum. It was withdrawn from the market in 1961 because of reports of congenital defects when taken during gestation. More recently, thalidomide has been shown to prevent neoangiogenesis in human malignancies and to exert immunomodulatory and anti-inflammatory properties, although the exact mechanism of action is unclear.1-5  Thalidomide was first used 10 years ago to treat patients with multiple myeloma (MM). This review addresses the role of thalidomide in the treatment of MM and the management of its side effects.

Thalidomide, a synthetic glutamic acid derivative (Figure 1), is poorly soluble in water, and thus no parenteral preparation is available. Thalidomide exhibits linear and dose-dependent pharmacokinetics that are not affected by age, sex, race, food, or smoking status. After a single dose ranging from 50 mg to 400 mg, plasma half-life is roughly 5.5 to 7.3 hours.6 

Figure 1

Molecular formula of thalidomide.

Figure 1

Molecular formula of thalidomide.

Close modal

Studies in people have shown little involvement of hepatic enzymes; thalidomide is minimally metabolized by the cytochrome P450 isoenzyme system,7  and interactions with drugs metabolized by this system are unlikely. Products of spontaneous hydrolysis are excreted in the urine. Most of an absorbed oral dose is eliminated through urine and feces.6 

Detectable concentrations of thalidomide have been noted in the semen and plasma of 2 of 4 seropositive patients with HIV given thalidomide at 100 mg/day for 8 weeks.8  The presence of thalidomide in semen is important because the risk for birth defects; thalidomide exposure in this context is unknown.

Thalidomide has been licensed for the treatment of relapsed-refractory MM in Australia, New Zealand, Turkey, Israel, and South Korea. The US Food and Drug Administration has approved thalidomide for the acute treatment of the cutaneous manifestation of erythema nodosum leprosum and, in combination with dexamethasone, for treatment of newly diagnosed patients with MM. Thalidomide was designated as an orphan medicinal product by the European Commission in 2001.

Single-agent thalidomide

Single-agent thalidomide was used by Singhal and colleagues almost 10 years ago.9  This crucial study showed the future development of the drug, since it first established the in vivo efficacy of thalidomide against myeloma. In relapsed patients, thalidomide monotherapy showed partial response (PR) rates of 25% to 30%. Most responses occurred within 2 months, and the median response duration was 12 months. The 2-year event-free (EFS) and overall survival (OS) were 20% and 48%, respectively.9-11  A systematic review of phase 2 trials identified 42 trials investigating 1674 patients. The PR rate was 29.4%. Minor response rate was 13.8%, and stable disease rate was 11%.12  Patients with extramedullary involvement had a lower response rate than did those without extramedullary disease (0% vs 59%; P < .001).13 

In a prospective randomized study,14  400 patients with relapsed-refractory MM received thalidomide at a dose of 100 mg/day or 400 mg/day, and no difference in 1-year OS was recorded (73% vs 69%, respectively). The 2 subgroups who received thalidomide at 100 mg/day or 400 mg/day in combination with dexamethasone also showed identical response rates. The 100 mg/day regimen was better tolerated and somnolence, constipation, and peripheral neuropathy were significantly reduced. The rate of deep-vein thrombosis (DVT) was much the same in both groups.14  These results accord with the use of lower doses of thalidomide in combination with dexamethasone.

Thalidomide monotherapy has been given to patients with smoldering or asymptomatic myeloma. Although responses have been reported, the approach is not recommended at this stage of disease, since some patients can be stable for several months or years without any therapy.15,16 

Advanced multiple myeloma

In various studies,17-19  the thalidomide-dexamethasone (TD) combination (Tables 1,2) induced PR rates ranging from 41% to 65%; the median time to response was lowered to 1 month or less. Hematologic side effects were uncommon; thus, patients with advanced disease and compromised bone marrow function could be treated. Constipation, somnolence, and neuropathy were the most frequent adverse effects. Neurologic side effects were the main reason for thalidomide dose reduction. Thromboembolic events have been reported in 2% to 7% of patients who did not receive any anticoagulant prophylaxis, but dexamethasone was mainly given at low doses (40 mg/day, 4 days/month; Table 3).17-19  In a case-control analysis,17  the efficacy of TD was compared with that of a control group who were given conventional chemotherapy. Results showed that TD was better than chemotherapy; median progression-free survival (PFS) was 17 months in the TD group and 11 months in the control group (P = .002). Three-year OS was 60% after TD and 26% after chemotherapy (P = .001).17  A randomized study20  confirmed the efficacy of the TD regimen compared with high-dose dexamethasone. TD induced higher PR rates (65% vs 28%; P < .001) and longer 1-year PFS (47% vs 31%; P = .009).

Table 1

Thalidomide in relapsed-refractory patients with MM

RegimenNo. patientsCR, %Greater than PR, %PFS/EFS/TTP, %OS, %Reference
Thalidomide alone       
    T 84 25 50 at 3 mo 58 at 12 mo 9  
    T 169 14* 30 20 at 24 mo 48 at 24 mo 10  
    T 400 ND ND ND 69-73 at 12 mo 14  
    T 1674 ND 29 50 at 12 mo 50 at 14 mo 12  
Dexamethasone       
    TD 62 ND 56 50 at 17 mo 60 at 36 mo 17  
    TD 58 ND 46 50 at 11 mo 50 at 19 mo 17  
    TD 77 ND 41 50 at 12 mo ND 18  
    TD 44 ND 55 50 at 4 mo 50 at 13 mo 19  
    TD 58 ND 65 47 at 12 mo ND 20  
Melphalan       
    TM 27 ND 59 61 at 24 mo 61 at 24 mo 21  
    MivPT 24 ND 41 50 at 9 mo 50 at 14 mo 22  
    MTD 21 18 50 at 9 mo 50 at 13 mo 23  
Doxorubicin       
    TAD 50 26 76 50 at 17 mo ND 24  
    DVd-T 49 20 75 50 at 16 mo 50 at 40 mo 25  
Cyclophosphamide       
    CDT 52 17 79 34 at 24 mo 73 at 24 mo 26  
    Pulsed CTD 53 60 50 at 8 mo 50 at 18 mo 27  
    ThaCyDex 71 55 57 at 24 mo 66 at 24 mo 28  
    HyperCTD 60 72 50 at 11 mo 50 at 19 mo 29  
RegimenNo. patientsCR, %Greater than PR, %PFS/EFS/TTP, %OS, %Reference
Thalidomide alone       
    T 84 25 50 at 3 mo 58 at 12 mo 9  
    T 169 14* 30 20 at 24 mo 48 at 24 mo 10  
    T 400 ND ND ND 69-73 at 12 mo 14  
    T 1674 ND 29 50 at 12 mo 50 at 14 mo 12  
Dexamethasone       
    TD 62 ND 56 50 at 17 mo 60 at 36 mo 17  
    TD 58 ND 46 50 at 11 mo 50 at 19 mo 17  
    TD 77 ND 41 50 at 12 mo ND 18  
    TD 44 ND 55 50 at 4 mo 50 at 13 mo 19  
    TD 58 ND 65 47 at 12 mo ND 20  
Melphalan       
    TM 27 ND 59 61 at 24 mo 61 at 24 mo 21  
    MivPT 24 ND 41 50 at 9 mo 50 at 14 mo 22  
    MTD 21 18 50 at 9 mo 50 at 13 mo 23  
Doxorubicin       
    TAD 50 26 76 50 at 17 mo ND 24  
    DVd-T 49 20 75 50 at 16 mo 50 at 40 mo 25  
Cyclophosphamide       
    CDT 52 17 79 34 at 24 mo 73 at 24 mo 26  
    Pulsed CTD 53 60 50 at 8 mo 50 at 18 mo 27  
    ThaCyDex 71 55 57 at 24 mo 66 at 24 mo 28  
    HyperCTD 60 72 50 at 11 mo 50 at 19 mo 29  

TTP indicates time to progression; T, thalidomide; TM, thalidomide-melphalan; MivPT, intravenous melphalan-thalidomide-prednisone; MTD, melphalan-thalidomide-dexamethasone; TAD, thalidomide-pegylated lyposomal doxorubicin-dexamethasone; DVd-T, pegylated lyposomal doxorubicin-vincristine-decreased frequency dexamethasone-thalidomide; CDT, cyclophosphamide-dexamethasone-thalidomide; CTD, cyclophosphamide-thalidomide-dexamethasone; ThaCyDex, thalidomide-cyclophosphamide–dexamethasone; and ND, not determined.

*

CR plus near CR.

Randomized trial.

Serum monoclonal protein reduction greater than 96%.

Table 2

Grades 3 to 4 toxic effects of thalidomide in relapsed-refractory patients with MM

RegimenNo. patientsThrombocytopenia, %Neutropenia, %Infection, %Peripheral neuropathy, %Reference
Thalidomide alone       
    T 84 ND ND ND ND 9  
    T 169 ND ND ND 9* 10  
    TD 1674 ND 11 ND 12  
Dexamethasone       
    TD 120 ND ND ND 17  
    TD 44 ND ND ND ND 19  
Melphalan       
    TM 27 ND 30 ND 40* 21  
    MivPT 24 19 37 22  
    MTD 21 38 52 ND 23  
Doxorubicin, TAD 50 16 16 24  
Cyclophosphamide       
    Pulsed CTD 53 26 ND 27  
    ThaCyDex 71 ND 10 6* 28  
    HyperCTD 60 30 86 26 16 29  
RegimenNo. patientsThrombocytopenia, %Neutropenia, %Infection, %Peripheral neuropathy, %Reference
Thalidomide alone       
    T 84 ND ND ND ND 9  
    T 169 ND ND ND 9* 10  
    TD 1674 ND 11 ND 12  
Dexamethasone       
    TD 120 ND ND ND 17  
    TD 44 ND ND ND ND 19  
Melphalan       
    TM 27 ND 30 ND 40* 21  
    MivPT 24 19 37 22  
    MTD 21 38 52 ND 23  
Doxorubicin, TAD 50 16 16 24  
Cyclophosphamide       
    Pulsed CTD 53 26 ND 27  
    ThaCyDex 71 ND 10 6* 28  
    HyperCTD 60 30 86 26 16 29  

ND indicates not determined.

*

Grades 2 to 4.

Leukopenia.

Grade 4 only.

Table 3

Thromboembolic events during thalidomide treatment

DVT/PE incidence without thromboprophylaxis
DVT/PE incidence with thromboprophylaxis
Newly diagnosed patients, %ReferenceRelapsed/refractory patients, %ReferenceFixed low-dose warfarin, %Full-dose warfarin, %Aspirin, %LMWH, %Reference
Thalidomide alone 30 2-3 10,12 ND ND ND ND — 
Thalidomide plus dexamethasone 12-26 31-34 2-7 17,19 12-25 ND ND 15,32,35,36  
Thalidomide plus melphalan 6-20 37-39 4-11 21,22 ND ND ND 37  
Thalidomide plus doxorubicin 10-27 40-42 58* 43 12-14 ND 14-18 5-9 24,43,,46  
Thalidomide plus cyclophosphamide 3*-11 47,48 2-8 27-29 12 ND ND ND 26  
Thalidomide plus multiagent chemotherapy 16-34 49,50 15 15 31 ND ND 15-24 49,51  
DVT/PE incidence without thromboprophylaxis
DVT/PE incidence with thromboprophylaxis
Newly diagnosed patients, %ReferenceRelapsed/refractory patients, %ReferenceFixed low-dose warfarin, %Full-dose warfarin, %Aspirin, %LMWH, %Reference
Thalidomide alone 30 2-3 10,12 ND ND ND ND — 
Thalidomide plus dexamethasone 12-26 31-34 2-7 17,19 12-25 ND ND 15,32,35,36  
Thalidomide plus melphalan 6-20 37-39 4-11 21,22 ND ND ND 37  
Thalidomide plus doxorubicin 10-27 40-42 58* 43 12-14 ND 14-18 5-9 24,43,,46  
Thalidomide plus cyclophosphamide 3*-11 47,48 2-8 27-29 12 ND ND ND 26  
Thalidomide plus multiagent chemotherapy 16-34 49,50 15 15 31 ND ND 15-24 49,51  

PE indicates pulmonary embolism; ND, not determined; and —, not applicable.

*

Includes newly diagnosed and relapsed patients.

Newly diagnosed MM

Two randomized studies have shown that the TD regimen is better than high-dose dexamethasone (Tables 4,5).31,52  Patients received thalidomide at a dose of 200 mg/day plus dexamethasone at a dose of 40 mg/day on days 1 to 4, 9 to 12, and 17 to 20 or dexamethasone alone at the same schedule. In the first study,31  the response rate was significantly higher with TD than with dexamethasone alone (63% vs 41%; P = .001). The rates of any grades 3 to 4 toxic effects were significantly higher with TD (45% vs 21%; P = .001). Both DVT (17% vs 3%) and grades 3 to 4 peripheral neuropathy (7% vs 4%) were more frequent in the TD group than in the dexamethasone group. The investigators concluded that lower doses of dexamethasone could greatly reduce the incidence of adverse events without a negative effect on efficacy, and that anticoagulant prophylaxis should be used in all newly diagnosed patients. No substantial differences in survival were reported.31  The second study showed that time to progression was also improved in the TD group (P < .001).52 

Table 4

Thalidomide in patients with newly diagnosed MM

RegimenNo. patientsCR, %Greater than PR, %PFS/EFS/TTP, %OS, %Reference
Dexamethasone       
    TD 235 49 50 at 17 mo ND 52 * 
    TD 103 63 50 at 22 mo 72 at 24 mo 31 * 
    TD 100 10 76 ND ND 32  
    TD 50 ND 64 ND ND 34  
    TD 100 ND 25 ND ND 33 * 
    TD 136 14 68 50 at 25 mo 50 at 45 mo 35 * 
Melphalan       
    MPT 129 16 76 54 at 24 mo 80 at 36 mo 37 * 
    MPT 125 13 76 50 at 28 mo 50 at 52 mo 38 * 
    MPT 113 62 50 at 24 mo 50 at 45 mo 39 * 
Doxorubicin       
    ThaDD 50 34 88 57 at 36 mo 74 at 36 mo 44  
    TAD 193 ND 79-81 ND ND 53 * 
    DVd-T 55 36 83 50 at 28 mo ND 25  
    T-VAD doxil 117 15 81 59 at 24 mo 77 at 24 mo 45 * 
    T-VAD doxil 39 10 74 55 at 22 mo 74 at 22 mo 40  
Cyclophosphamide, CTD 27 15 89 ND ND 47  
RegimenNo. patientsCR, %Greater than PR, %PFS/EFS/TTP, %OS, %Reference
Dexamethasone       
    TD 235 49 50 at 17 mo ND 52 * 
    TD 103 63 50 at 22 mo 72 at 24 mo 31 * 
    TD 100 10 76 ND ND 32  
    TD 50 ND 64 ND ND 34  
    TD 100 ND 25 ND ND 33 * 
    TD 136 14 68 50 at 25 mo 50 at 45 mo 35 * 
Melphalan       
    MPT 129 16 76 54 at 24 mo 80 at 36 mo 37 * 
    MPT 125 13 76 50 at 28 mo 50 at 52 mo 38 * 
    MPT 113 62 50 at 24 mo 50 at 45 mo 39 * 
Doxorubicin       
    ThaDD 50 34 88 57 at 36 mo 74 at 36 mo 44  
    TAD 193 ND 79-81 ND ND 53 * 
    DVd-T 55 36 83 50 at 28 mo ND 25  
    T-VAD doxil 117 15 81 59 at 24 mo 77 at 24 mo 45 * 
    T-VAD doxil 39 10 74 55 at 22 mo 74 at 22 mo 40  
Cyclophosphamide, CTD 27 15 89 ND ND 47  

ThaDD indicates thalidomide-pegylated liposomal doxorubicin-dexamethasone; T-VAD doxil, thalidomide-vincristine-pegylated lyposomal doxorubicin; and ND, not determined.

*

Randomized trial.

Updated information was presented at the American Society of Clinical Oncology and European Hematology Association Congress.

Very good partial response rate.

Table 5

Grades 3 to 4 toxic effects of thalidomide in patients with newly diagnosed MM

RegimenNo. patientsThrombocytopenia, %Neutropenia, %Infection, %Peripheral neuropathy, %Reference
Dexamethasone       
    TD 235 ND 52 * 
    TD 103 ND ND 31 * 
    TD 100 ND 32  
    TD 50 ND ND ND 30  
    TD 100 ND ND ND ND 33 * 
Melphalan       
    MPT 129 16 10 37 * 
    MPT 125 14 48 13 38 * 
    MPT 113 ND 23 ND 20 39 * 
Doxorubicin       
    ThaDD 50 12 22 44  
    T-VAD doxil 117 8 10 6 45 * 
    T-VAD doxil 39 15 15 21 40  
Cyclophosphamide, CTD 27 ND 47  
RegimenNo. patientsThrombocytopenia, %Neutropenia, %Infection, %Peripheral neuropathy, %Reference
Dexamethasone       
    TD 235 ND 52 * 
    TD 103 ND ND 31 * 
    TD 100 ND 32  
    TD 50 ND ND ND 30  
    TD 100 ND ND ND ND 33 * 
Melphalan       
    MPT 129 16 10 37 * 
    MPT 125 14 48 13 38 * 
    MPT 113 ND 23 ND 20 39 * 
Doxorubicin       
    ThaDD 50 12 22 44  
    T-VAD doxil 117 8 10 6 45 * 
    T-VAD doxil 39 15 15 21 40  
Cyclophosphamide, CTD 27 ND 47  

ND indicates not determined.

*

Randomized trial.

Updated information was presented at the American Society of Clinical Oncology and European Haematology Association Congress.

Grades 2 to 4.

In a continuing study,35  274 elderly patients were randomly assigned to TD or oral melphalan and prednisone (MP). Thalidomide was given at 200 to 400 mg/day, and dexamethasone was given at 40 mg/day on days 1 to 4 and 15 to 18 on odd cycles, and on days 1 to 4 on even cycles. TD induced higher PR rates than MP (68% vs 51%; P = .004). In patients older than 72 years, TD induced a higher mortality rate and a worse OS than did MP (median, 45 months vs 58 months; P = .029). Overall survival was much the same in patients younger than 72 years (58 months vs 50 months; P = .186). DVT, neuropathy, and constipation were more frequent in the TD group, whereas hematologic toxic effects were most prominent in the MP group.35 

A case-control analysis showed that the TD regimen was better than the vincristine-doxorubicin-dexamethasone (VAD) schema.32  Adverse events were more frequent in the TD group; the DVT rate was 26% in the first 19 patients who did not receive any anticoagulant prophylaxis and 12% in the subsequent 81 patients who received low-dose warfarin as prophylaxis. A prospective randomized study33  compared TD with VAD. The frequency of at least very good partial response (VGPR) was 25% after TD induction compared with 7% after VAD (P = .003). However, the benefit of TD was not confirmed at 6 months after autologous stem- cell transplantation (ASCT), since VGPR rates (44% vs 42%; P = .87) were almost identical. DVT was higher in the TD group (23% vs 8%; P = .004).

Advanced multiple myeloma

Offidani and coworkers21  showed that oral melphalan and thalidomide was better than thalidomide alone (Tables 1,2). Thalidomide has also been associated with low-dose intravenous melphalan (20 mg/m2 every 4 months) in one study,22  and with intermediate-dose intravenous melphalan in another.23  Hematologic toxic effects were manageable in the first study,21  but were quite severe in the other 2 studies.22,23 

Newly diagnosed MM

A total of 3 randomized studies assessed the combination of MP and thalidomide (Tables 4,5). In Palumbo et al's study,37  the oral standard MP was compared with MP plus thalidomide (MPT) in patients aged 60 to 85 years. The PR rates were 76.0% in the MPT group and 47.6% in the MP group. The 2-year EFS was 54% after MPT and 27% after MP (P < .001), and 3-year OS was 80% for MPT and 64% for MP (P = .19). The incidence of grades 3 to 4 adverse events was significantly higher in the MPT group than in the MP group (48% vs 25%; P < .001); most frequent were hematological toxic effects (22%), thromboembolism (12%), infections (10%), and peripheral neuropathy (8%). After the introduction of prophylactic enoxaparin, the frequency of DVT was lowered from 20% to 3% (P = .005).

Facon and colleagues38  compared MP with both MPT and ASCT (melphalan, 100 mg/m2) in patients aged 65 to 75 years. PR rates were 35%, 76%, and 65% (P < .001), and median PFSs were 17.8, 27.5, and 19.4 months, respectively; PFS was significantly longer in the MPT group than in the MP (P < .001) and ASCT (P < .001) groups, but no difference was noted between the MP and ASCT groups (P = .25). Median OS was 33.2, 51.6, and 38.3 months in the MP, MPT, and ASCT groups, respectively. Similarly, OS was significantly extended after MPT (P < .001) and ASCT (P = .027), although investigators noted no difference between the MP and ASCT groups (P = .86). The most frequent side effects were neutropenia, thrombocytopenia, infections, constipation, and peripheral neuropathy; thromboembolic events were reported in 12% of patients, but no prophylaxis was adopted.

In Hulin et al's trial,39  patients aged 75 years and older were randomly assigned to MP or MPT. The PR rate was higher in the MPT group than in the MP group (62% vs 31%), and both median PFS (24 months vs 19 months; P = .001) and OS (45 months vs 28 months; P = .03) were significantly longer. Toxic effects were higher in patients assigned to MPT and were mainly because of peripheral neuropathy (20%), neutropenia (23%), and depression (7%); no significant differences in DVT rate between the 2 groups were noted (6% vs 4%).

These 3 independent randomized studies showed virtually identical results, and they represent the experimental evidence that supports MPT as the standard of care for elderly patients not eligible for ASCT.

Advanced MM

Several trials have investigated the combination of thalidomide, pegylated lyposomal doxorubicin (PLD), and dexamethasone (Tables 1,2). Offidani and coworkers24  used a VAD-like regimen that substituted vincristine with low-dose thalidomide. Hussein and colleagues25  added thalidomide to a regimen consisting of PLD, vincristine, and decreased-frequency dexamethasone. Complete response (CR) and PR rates were similar between regimens; the toxic effects of these regimens were manageable, although vincristine increased the incidence of severe neuropathy.

Newly diagnosed MM

The association of thalidomide, doxorubicin, and dexamethasone (TAD) has been studied in newly diagnosed patients (Tables 4,5). In a phase 2 study,44  the PR rate was 88%, including 34% CR. Patients with impaired renal function achieved PR, and renal function was improved or restored. The rate of DVT was 14% despite the use of low-dose warfarin. Severe infections were reported in 22% patients, although the introduction of antibiotic prophylaxis decreased the frequency of such adverse events. In 2 parallel phase 3 multicenter German-Dutch studies,53  newly diagnosed patients with MM were randomly assigned to received TAD or the standard VAD regimen as induction treatment before ASCT. An interim analysis showed a better PR rate with TAD than with VAD both in the German study (79% vs 58%) and in the Dutch trial (81% vs 61%). Patients treated with TAD had fewer peripheral blood stem cells; nevertheless, the stem cells obtained were sufficient for double ASCT in 82% of patients in the TAD group.

A randomized trial compared VAD-doxil plus thalidomide (TVAD-doxil) with VAD-doxil.45  Partial response rates were more frequent in the TVAD-doxil group than in the VAD-doxil group (81% vs 63%; P = .003); extended 2-year PFS (59% vs 45%; P = .013) and increased OS (77% vs 64%; P = .037) were reported for patients assigned to TVAD-doxil.

Other studies used vincristine and PLD in association with thalidomide (400 mg) and decreased-frequency dexamethasone,25  or with thalidomide (200 mg) and high-dose dexamethasone.40  The PR rate ranged from 74% to 83%, and major toxic effects included DVT, peripheral neuropathy, palmar-plantar erythrodisestesia, and infections.

Advanced MM

Cyclophosphamide has been associated with thalidomide and corticosteroids for the treatment of advanced MM (Tables 1,2). Oral weekly cyclophosphamide (300 mg/m2 per week), monthly pulsed low-dose dexamethasone, and thalidomide (300 mg/day; CDT) has been studied. The PR rate was 62%, including 17% CR. A total of 27 percent of patients developed infections, most of which were respiratory, but there were no infection-related deaths.26 

In another phase 2 study,27  patients received pulsed cyclophosphamide (150 mg/m2 twice daily, days 1-5), thalidomide (400 mg, days 1-5 and 14-18) and dexamethasone (20 mg/m2, days 1-5 and 14-18). Toxic effects were mild or moderate, and the cumulative frequencies of DVT and peripheral neuropathy were 2% and 4%. Other phase 2 studies showed similar results.28,29 

Newly diagnosed MM

The combinations of oral cyclophosphamide (500 mg on days 1, 8, and 15), thalidomide (400 mg), and high-dose dexamethasone (CTD) have been studied in 27 patients and compared with the combination of cyclophosphamide-vincristine-doxorubicin-methylprednisolone (CVAMP) in a case-matched analysis (Tables 4,5). The PR rate was significantly higher with CTD than with CVAMP (89% vs 56%; P = .016). The frequency of DVT was higher in the CTD group (11% vs 4%), but grades 3 to 4 neutropenia (4% vs 60%) and infections (7% vs 26%) were greater in the CVAMP group.47 

To assess whether the incorporation of thalidomide into high-dose therapy could improve survival, 668 newly diagnosed patients, who received tandem ASCT, were randomly assigned to receive thalidomide or not.49  Patients given thalidomide had a higher CR rate (62% vs 43%; P < .001) and a higher 5-year EFS rate (56% vs 44%; P = .01). There was no difference in OS because of a significantly shorter survival after relapse in the thalidomide group (median, 1.1 years vs 2.7 years; P = .001). Of note, 83% patients in the control group received thalidomide at relapse. The delivery of thalidomide at diagnosis significantly extended EFS, but the administration of thalidomide at relapse in the control group probably abrogated the OS benefit. Thromboembolic events were more common in the thalidomide group (34% vs 18%; P < .001). The high rate of DVT was not prevented by the introduction later in the study of prophylactic low-molecular-weight heparin (LMWH; 24% in the thalidomide group vs 15% in the control group; P = .064). Peripheral neuropathy (grade 2 or higher) was more common in the thalidomide group (27% vs 17%; P < .001). Syncopal episodes related to sinus bradycardia arose in 12% of patients in the thalidomide group (4% in the control group). Cardiac pacemakers were implanted in nearly a third of patients with symptomatic sinus bradycardia. The treatment-related mortality rate was similar in both groups.49 

High-dose chemotherapy has increased the response rate in patients with MM, but this treatment is not curative, and effective consolidation-maintenance strategies could extend the duration of response.

In a large study,54  2 months after ASCT, patients were randomly assigned to receive no maintenance (group A), pamidronate (group B), or pamidronate plus thalidomide (group C; 400 mg/day, dose reduction to a minimum dose of 50 mg was allowed for treatment-related toxicity). At least VGPR was achieved in 55% of patients (group A), 57% of patients (group B), and 67% of patients (group C; P = .03). The 3-year EFS was 36% (group A), 37% (group B), and 52% (group C; P < .009). The 4-year OS was 77% (group A), 74% (group B), and 87% (group C; P < .04). Patients received thalidomide for a median of 15 months (range, 0.1-50 months). Drug-related adverse events led to discontinuation of thalidomide in 39% of patients, mainly because of peripheral neuropathy. The mean dose of thalidomide was 200 mg/day. Most frequent toxic effects included neuropathy (68%), fatigue (34%), constipation (20%), neutropenia (7%), and cardiac events (4%). The DVT incidence did not differ significantly in the 3 groups.

In an Australian randomized study,55  thalidomide (200 mg/day for a maximum of 12 months) combined with prednisolone (50 mg on alternate days) was compared with prednisolone alone as maintenance after ASCT. At 12 months after randomization, the thalidomide group had higher PR rate (83% vs 52%; P < .01), better 2-year PFS (63% vs 36%; P < .001), and better 3-year OS (86% vs 75%; P = .02).

In a recent study, patients with newly diagnosed MM were randomly assigned to receive either a single ASCT followed by thalidomide maintenance or tandem ASCT. Single ASCT followed by maintenance with thalidomide proved better than tandem ASCT for both 3-year PFS (85% vs 57%; P = .02) and 3-year OS (85% vs 65%; P = .04).56  Whether the most effective strategy is maintenance treatment with thalidomide or therapy with thalidomide at relapse is still debated.

Factors affecting outcome of patients with MM treated with thalidomide have been assessed. These studies might select patients who most benefit from thalidomide and might allow a risk-based treatment algorithm to be designed.

In the first study into the use of thalidomide monotherapy in advanced MM,9  3 factors were associated with a short EFS: increased LDH, C-reactive protein, and plasma cell labeling index. Short OS was associated with low albumin, chromosome 13 deletion (del13), and high bone marrow plasma cell infiltration. Barlogie and colleagues49  treated patients with ASCT with or without thalidomide. In the thalidomide group, EFS was independent of cytogenetics abnormalities, high LDH, or low serum albumin. Thalidomide was unable to overcome these adverse prognostic factors in terms of OS. The Bologna group57  reported a significantly reduced probability of response to TD in patients with coexisting del13 or t(4;14) cytogenetics abnormalities, but not in those with a single abnormality. These negative results were completely offset by subsequent tandem ASCT.57  In another study of relapsed patients given TD,58  time to first progression of less than 12 months was significantly associated with shorter OS (P = .002), but serum elevated β2-microglobulin and C-reactive protein did not predict poor outcome. Attal and coworkers54  showed that patients without del13 had a significant benefit from thalidomide, whereas patients with del13 did not.

The frequency and severity of thalidomide side effects are dose related and time dependent (Tables 2,3,5). Physicians should grade side effects according to the National Cancer Institute Common Toxicity Criteria for Adverse Events. Generally, once a relation between thalidomide and toxicity is established, no action should be taken if there are nonhematologic grade 1 side effects. If there are grade 2 toxic effects, the thalidomide dose should be reduced to 50%; for grades 3 to 4, it should be discontinued.

Teratogenicity

Birth defects were observed in 10 000 to 12 000 infants before thalidomide was withdrawn.59  Thalidomide does not seem to be carcinogenic, genotoxic, or mutagenic, and it has not been correlated to second-generation birth defects.60,61  Thalidomide administration in pregnant women is absolutely contraindicated. Women of childbearing potential should have a negative pregnancy test before starting thalidomide, use 2 effective forms of birth control, and have a pregnancy test every 4 weeks if their period is regular or every 2 weeks in case of irregularity. Men receiving thalidomide must abstain from sexual intercourse or use a latex condom even if they have undergone vasectomy. The Risk Management Programs are designed to avoid prescription of thalidomide to pregnant women or men with childbearing potential.

Thromboembolism

Thromboembolic events are one of the most important side effects (Table 3). The pathogenic mechanisms of DVT associated with thalidomide have not been clearly established. Acquired activated protein C resistance and a reduction in thrombomodulin level have been associated with an increased risk of DVT.62,63  No specific coagulation test can predict the high risk of DVT. Endothelial injury produced by the combination of thalidomide with chemotherapy and subsequent restoration of endothelial cell PAR-1 expression are probably factors that promote thrombosis.64  Since thalidomide does not cause endothelial damage,65  the frequency of DVT is low when thalidomide is used as monotherapy, whereas it substantially increases when chemotherapy is added.

The goal of thromboprophylaxis should be to reduce the frequency of DVT to less than 10%. Thromboprophylaxis is not recommended for patients who receive single-agent thalidomide or thalidomide maintenance (if no other risk factors for DVT are simultaneously present), but it is strongly recommended in those who receive thalidomide in combination with high-dose dexamethasone or doxorubicin. Thromboprophylaxis should be tailored according to the presence of other risk factors that could further increase the risk of DVT: individual risk factors (age, obesity, comorbidities, surgical procedures), MM-related risk factors (diagnosis, high tumor mass), and treatment-related risk factors (high-dose dexamethasone, doxorubicin, multiagent chemotherapies). There are no data to suggest that one anticoagulant is better than another. Although aspirin is more appealing because of convenience and of ease of administration, the rate of thrombosis is relatively high in particular conditions. Until further evidence becomes available, aspirin should only be recommended for patients with low risk of DVT. In high-risk patients, evidence nowadays supports the use of LMWH or full-dose warfarin. The use of warfarin is limited by its long half-life in patients at risk of concomitant thrombocytopenia. LMWH is a more suitable option because of its short half-life and the decreased risk of secondary bleeding. Ongoing studies will determine which is the most appropriate anticoagulant prophylaxis.

Almost all DVT has been reported in the first 6 months of treatment, and all episodes arose within the first 12 months.37,50,51  Anticoagulant prophylaxis should be delivered for 6 months or for longer periods in the presence of additional risk factors.

Cardiovascular side effects

Sinus bradycardia occurs in patients given thalidomide because of autonomic neuropathy. Bradycardia can be severe enough to cause syncope.66  Patients should be assessed at the start of treatment to identify previous episodes such as syncope that can be associated with heart disease, especially when thalidomide is used in combination with chemotherapy such as doxorubicin or cyclophosphamide. Symptoms related to bradycardia can resolve or decrease in severity after either discontinuation or dose reduction, but sometimes a pacemaker needs to be implanted.49,67 

Hypotension is a possible although infrequent side effect. The concurrent administration of antidepressants could induce or worsen hypotension. The risks of hypotension and bradycardia increase in elderly patients receiving concomitant antihypertensive drugs such as β-blockers. Peripheral edema is more frequent with coadministration of corticosteroids; it is generally mild, reversible, and usually responsive to temporary discontinuation of the drug.66 

Neurologic side effects

Many patients with MM present with subclinical or even clinical peripheral neuropathy. These patients can be at increased risk of peripheral neuropathy because of thalidomide treatment. These effects usually arise after prolonged administration of thalidomide (70% of patients treated for 12 months, although effects are mainly grades 1-2).68  Clinical manifestations include sensory symptoms (distal paresthesia and hyperesthesia), motor symptoms, and autonomic dysfunction. Patients suffer from pinprick sensations, numbness, and tingling that initially affect fingers and toes but later extend proximally. Subsequently, vibration perception, deep sensitivity, and position sense are affected, leading to ataxia, progressive gait disturbance, and postural tremor. Neuropathy is closely related to duration of treatment68,69  and cumulative dose,70,71  and it is more frequent in elderly patients.

A randomized study17  reported a greater tolerability of thalidomide in patients who received 100 mg than in those who received 400 mg. Doses of 200 mg/day or less should be considered to keep side effects to a minimum; lower doses (100 mg/day) are better tolerated in elderly patients. Thalidomide discontinuation increases the probability of recovery, which usually occurs within 3 weeks. If treatment is not stopped, the neuropathy progresses and could become irreversible. Patients should be taught to recognize peripheral neuropathy and to immediately decrease the dose or discontinue thalidomide when sensory paresthesia is complicated by pain, motor deficit, or interference with daily function. A practical rule is to maintain the assigned dose if neuropathy is grade 1, to reduce the dose by 50% if neuropathy is grade 2, to discontinue the drug if neuropathy is grade 3, and to eventually resume thalidomide at a decreased dose if neuropathy reaches grade 1. Symptomatic therapy for paresthesia includes gabapentin, pregabalin, L-carnitine, and tricyclic antidepressants.

Tremors arise quite frequently but rarely interfere with daily activity. Depression has been reported, especially in patients with a previous history of depression. Headache is possible, it is not usually dose-dependent and can be relieved with nonsteroidal anti-inflammatory drugs.66 

Constipation

Constipation is the most common gastrointestinal side effect (more than 50% of patients), and it seems to be dose dependent. This adverse event appears early after treatment is started (2-4 days), and is most severe in elderly patients and in those receiving opioid analgesics.66  Patients should maintain a high fluid intake, a high fiber diet, and sufficient exercise. Drugs that reduce bowel motility should be avoided if possible. Stool softeners and osmotic laxative are usually sufficient for moderate constipation. For severe constipation, a 50% dose reduction is needed.

Sedation

Some amount of sedation is universal. It generally appears within 15 days after thalidomide treatment is started and is usually mild and dose dependent. Benzodiazepines, opiates, phenytoin, or sedative neuroleptics can enhance sedation, and thalidomide itself can increase the sedative activity of barbiturates and alcohol, and the catatonic activity of chlorpromazine and reserpine.66  Thalidomide does not produce central nervous system (CNS) or respiratory depression.59 

Patients should be instructed to take thalidomide in the evening and to expect assumption in late afternoon if somnolence persists the following morning. Cautions should be given in the first 2 weeks for activities requiring great attention (eg, driving), and concomitant drugs with sedative activity should be avoided.

Dermatologic side effects

Adverse dermatologic effects include rash, atrophic lesions, dry skin and mouth, and, rarely, toxic epidermic necrolysis and the Stevens-Johnson syndrome. The most common dermatologic side effect is a pruritic maculopapular rash, starting on the trunk and extending to the back and proximal limbs; it does not seem to be dose related and has been reported within 10 to 14 days after the start of treatment. Vesicular eruptions, usually pruritic, occur in 25% of patients given doses greater than 400 mg/day after 30 to 60 days of therapy. Temporary discontinuation leads to resolution of the rash and allows readministration at a reduced dose.

The most serious hypersensitivity reaction is toxic epidermic necrolysis, associated with mortality rates greater than 30%. The appearance of a maculopapular rash that covers the entire body is a potential early sign.72  Because of these possible dermatologic effects, coadministration of thalidomide with sulphonamides, allopurinol, or cotrimoxazole should be undertaken with caution.73 

Thyroid dysfunction

Thalidomide can rarely be thyrotoxic or provoke an immune reaction against the thyroid gland.66  Evidence of reduction in thyroid function, most of which is subclinical, has been reported from 2% to 35%.29,74  Subclinical hypothyroidism might contribute to the adverse effect profile of thalidomide, especially constipation, fatigue, neuropathy, skin rush, and bradycardia. Measurements of thyroid-stimulating hormone levels at baseline and at regular intervals (every 3 months) could detect subclinical hypothyroidism. Low-dose thyroxin supplementation should be given for cases of clinical and subclinical hypothyroidism.

Hematologic side effects and infections

Thalidomide can induce neutropenia, usually mild, in 3% to 15% of patients.9,75  The frequency increases when thalidomide is combined with myelotoxic chemotherapy. Thrombocytopenia is rare and generally associated with neutropenia. Impairment of marrow function is not a contraindication but rather an argument for the use of thalidomide and corticosteroids, since this combination does not induce substantial hematologic toxic effects.

Infections are an important complication: immunosuppression, exacerbated by the concomitant use of corticosteroids, is probably the main cause. Grades 3 to 4 infections were reported in 16% to 22% of patients given thalidomide, dexamethasone, and doxorubicin. Most infectious complications occurred during the first 3 courses of treatment; prophylaxis with ciprofloxacin reduced the infection rate to less than 10%.24,44 

Liver and renal function

Thalidomide-associated hepatitis, resolved after drug withdrawal, has been reported, although it is rare.76  Since thalidomide is spontaneously and nonspecifically hydrolyzed, plasma concentrations should not change in patients with compromised liver function. No data suggest that patients with damaged liver function need a dose reduction. Regular monitoring of liver enzymes (ie, during the first months of therapy and every 3 months thereafter) might be reasonable.

Renal adverse effects are rare. In patients with renal impairment, pharmacokinetics of thalidomide was similar to that reported in those with normal renal function.77  These data suggest that a thalidomide dose adjustment is not needed in patients with renal impairment. In patients with renal impairment (defined as serum creatinine greater than 130 mM/L), the frequency of side effects was comparable to that observed in patients with normal renal function.78 

After 40 years, thalidomide is the first new drug that yields high response rates and improves remission duration and survival for patients with MM.

TD is a new induction therapy for patients eligible for ASCT for whom stem-cell harvest is efficient. The response rate is better than it is with VAD before high-dose melphalan, but almost identical after ASCT. EFS data are needed before the benefit of this approach can be clearly established. Preliminary data suggest that thalidomide combinations including doxorubicin or cyclophosphamide might be more effective than TD. Maintenance treatment with low-dose thalidomide should be at least offered to patients not achieving VGPR after tandem ASCT. In elderly patients, MPT is now regarded as the standard of care after 3 independent randomized studies37-39  showed that MPT was better than the standard MP regimen. Combinations including thalidomide with dexamethasone, melphalan, doxorubicin, or cyclophosphamide are effective options for the management of relapsed-refractory disease.

An improved ratio of efficacy to side effects can be obtained when low-dose thalidomide is combined with steroids or chemotherapy. Somnolence and constipation are clearly dose dependent; DVT is a major side effect when thalidomide is used in combination with other drugs. Antithrombotic prophylaxis is mandatory in newly diagnosed patients for at least the first 6 months of therapy. Peripheral neuropathy is dose and time dependent; although mild symptoms might be acceptable, more severe symptoms (greater than grade 1) need a thalidomide dose reduction and discontinuation. Thalidomide can be safely given to patients with renal impairment.

In the future, the main issues will be to define the subgroups of patients who might benefit most from thalidomide-based regimens and to explore new combinations including bortezomib or lenalidomide. Both agents can be given to patients with established resistance to thalidomide. Lenalidomide showed little degree of cross-resistance with thalidomide, and bortezomib can have some restrictions in patients with preexisting thalidomide-related neuropathy. Whether these new combinations will replace ASCT or whether they should be incorporated to further improve the efficacy of standard ASCT is unknown. Finally, further studies should clarify whether tumor eradication rather than maintaining of an asymptomatic disease is the most effective strategy for MM.

We thank Tiziana Marangon for her technical assistance in the preparation of the manuscript.

Contribution: A.P., T.F., P.S., J.B., P.M., A.W., A.S., H.L., M.B., and J.-L.H. revised material and wrote the paper; and M.O. and F.G. collected data, revised material, and wrote the paper.

Conflict-of-interest disclosure: A.P., T.F., and M.B. have received scientific advisor board and lecture fees from Pharmion, Celgene, and Janssen-Cilag. P.S. has received scientific advisor board and lecture fees from Celgene. P.M. received honoraria from Pharmion in 2007. A.W. is a member of the Advisory Board, Nordic branch of Pharmion Speakers Bureau, Janssen-Cilag. J.-L.H. is a member of the Speakers Bureau and Advisory Board of Orthobiotech, Pharmion, and Celgene. All other authors declare no competing financial interests.

Correspondence: Antonio Palumbo, Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S Giovanni Battista, Via Genova 3, 10126 Torino, Italy; e-mail: appalumbo@yahoo.com.

1
Zwingenberger
 
K
Wnendt
 
S
Immunomodulation by thalidomide: systemic review of the literature and of unpublished observations.
J Inflamm
1996
, vol. 
46
 (pg. 
177
-
211
)
2
Hallek
 
M
Leif Bergsagel
 
P
Anderson
 
KC
Multiple myeloma: increasing evidence for a multistep transformation process.
Blood
1998
, vol. 
91
 (pg. 
3
-
21
)
3
Damiano
 
JS
Cress
 
AE
Hazlehurst
 
LA
, et al. 
Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines.
Blood
1999
, vol. 
93
 (pg. 
1658
-
1667
)
4
Bertolini
 
F
Mingrone
 
W
Alietti
 
A
, et al. 
Thalidomide in multiple myeloma, myelodysplastic syndromes and histiocytosis: analysis of clinical results and of surrogate angiogenesis markers.
Ann Oncol
2001
, vol. 
12
 (pg. 
987
-
990
)
5
Zangari
 
M
Elice
 
F
Tricot
 
G
Immunomodulatory drugs in multiple myeloma.
Expert Opin Investig Drugs
2005
, vol. 
14
 (pg. 
1411
-
1418
)
6
Teo
 
SK
Colburn
 
WA
Tracewell
 
WG
, et al. 
Clinical pharmacokinetics of thalidomide.
Clin Pharmacokinet
2004
, vol. 
43
 (pg. 
311
-
327
)
7
Teo
 
S
Sabourin
 
P
O'Brien
 
K
, et al. 
Metabolism of thalidomide in human microsomes, cloned human cytochrome P-450 isozymes and Hansen's disease patients.
J Biochem Mol Toxicol
2000
, vol. 
14
 (pg. 
140
-
147
)
8
Teo
 
SK
Harden
 
JL
Burke
 
AB
, et al. 
Thalidomide is distributed into human semen after oral dosing.
Drug Metab Dispos
2001
, vol. 
29
 (pg. 
1355
-
1357
)
9
Singhal
 
S
Mehta
 
J
Desikan
 
R
, et al. 
Antitumor activity of thalidomide in refractory multiple myeloma.
N Engl J Med
1999
, vol. 
341
 (pg. 
1565
-
1571
)
10
Barlogie
 
B
Desikan
 
R
Eddlemor
 
P
, et al. 
Extented survival in advanced and refractory multiple myeloma after single-agent thalidomide: identification of prognostic factors in a phase 2 study of 169 patients.
Blood
2001
, vol. 
98
 (pg. 
492
-
494
)
11
Cibeira
 
MT
Rosinol
 
L
Ramiro
 
L
, et al. 
Long-term results of thalidomide in refractory and relapsed multiple myeloma with emphasis on response duration.
Eur J Haematol
2006
, vol. 
77
 (pg. 
486
-
492
)
12
Glasmacher
 
A
Hahn
 
A
Hoffmann
 
F
, et al. 
A systematic review of phase II trials of thalidomide monotherapy in patients with relapsed or refractory multiple myeloma.
Br J Hematol
2005
, vol. 
132
 (pg. 
584
-
593
)
13
Rosinol
 
L
Cibeira
 
MT
Blade
 
J
, et al. 
Extramedullary multiple myeloma escapes the effect of thalidomide.
Haematologica
2004
, vol. 
89
 (pg. 
832
-
836
)
14
Yakoub-Agha
 
I
Doyen
 
C
Hulin
 
C
, et al. 
A multicenter prospective randomised study testing non inferiority of thalidomide 100 mg/day as compared with 400 mg/day in patients with refractory/relapsed multiple myeloma: results of the final analysis of the IFM 01–02 study [abstract].
J Clin Oncol
2006
, vol. 
24
 pg. 
7520
 
15
Weber
 
D
Rankin
 
K
Gavino
 
M
, et al. 
Thalidomide alone or with dexamethasone for previously untreated multiple myeloma.
J Clin Oncol
2003
, vol. 
21
 (pg. 
16
-
19
)
16
Rajkumar
 
SV
Dispenzieri
 
A
Fonseca
 
R
, et al. 
Thalidomide for previously untreated indolent or smoldering multiple myeloma.
Leukemia
2001
, vol. 
15
 (pg. 
1274
-
1276
)
17
Palumbo
 
A
Bertola
 
A
Falco
 
P
, et al. 
Efficacy of low dose thalidomide as first salvage regimen in multiple myeloma.
Hemat J
2004
, vol. 
5
 (pg. 
318
-
324
)
18
Palumbo
 
A
Giaccone
 
L
Bertola
 
A
, et al. 
Low-dose thalidomide plus dexamethasone is an effective therapy for advanced myeloma.
Haematologica
2001
, vol. 
86
 (pg. 
399
-
403
)
19
Dimopoulos
 
MA
Zervas
 
K
Kouvatseas
 
G
, et al. 
Thalidomide and dexamethasone combination or refractory multiple myeloma.
Ann Oncol
2001
, vol. 
12
 (pg. 
991
-
995
)
20
Fermand
 
JP
Jaccard
 
A
Macro
 
M
, et al. 
A randomized comparison of dexamethasone + thalidomide (Dex/Tal) vs Dex + Placebo (Dex/P) in patients (pts) with relapsing multiple myeloma (MM) [abstract].
Blood
2006
, vol. 
108
  
Abstract 3563
21
Offidani
 
M
Corvatta
 
L
Marconi
 
M
, et al. 
Thalidomide plus oral melphalan compared with thalidomide alone for advanced multiple myeloma.
Hematol J
2004
, vol. 
5
 (pg. 
312
-
317
)
22
Palumbo
 
A
Avonto
 
I
Bruno
 
B
, et al. 
Intravenous melphalan, thalidomide and prednisone in refractory and relapsed multiple myeloma.
Eur J Hematol
2006
, vol. 
76
 (pg. 
273
-
277
)
23
Srakovic
 
G
Elson
 
P
Trebisky
 
B
, et al. 
Use of melphalan, thalidomide and dexamethasone in treatment of refractory and relapsed multiple myeloma.
Med Oncol
2002
, vol. 
19
 (pg. 
219
-
226
)
24
Offidani
 
M
Corvatta
 
L
Marconi
 
M
, et al. 
Low-dose thalidomide with pegylated liposomal doxorubicin and high-dose dexamethasone for relapsed/refractory multiple myeloma: a prospective, multicenter, phase II study.
Haematologica
2006
, vol. 
91
 (pg. 
133
-
136
)
25
Hussein
 
MA
Baz
 
R
Srkalovic
 
G
, et al. 
Phase 2 study of pegylated liposomal doxorubicin, vincristine, decreased-frequency dexamethasone, and thalidomide in newly diagnosed and relapsed-refractory multiple myeloma.
Mayo Clin Proc
2006
, vol. 
81
 (pg. 
889
-
895
)
26
Kyriakou
 
C
Thomson
 
K
D'Sa
 
S
, et al. 
Low dose thalidomide in combination with oral weekly cyclophosphamide and pulsed dexamethasone is a well tolerated and effective regimen in patients with relapsed and refractory multiple myeloma.
Br J Hematol
2005
, vol. 
129
 (pg. 
763
-
770
)
27
Dimopoulos
 
MA
Hamilos
 
G
Zomas
 
A
, et al. 
Pulsed cyclophosphamide, thalidomide and dexamethasone: an oral regiment for previously treated patients with multiple myeloma.
Hematol J
2004
, vol. 
5
 (pg. 
112
-
117
)
28
Garcia-Sanz
 
R
Gonzalez-Porras
 
JR
Hernández
 
JM
, et al. 
The oral combination of thalidomide, cyclophosphamide and dexamethasone is effective in relapsed/refractory multiple myeloma.
Leukemia
2004
, vol. 
18
 (pg. 
856
-
863
)
29
Kropff
 
MH
Lang
 
N
Bisping
 
G
, et al. 
Hyperfractionated cyclophosphamide in combination with pulsed dexamethasone and thalidomide (HyperCTD) in primary refractory or relapsed multiple myeloma.
Br J Haematol
2003
, vol. 
122
 (pg. 
607
-
616
)
30
Rajkumar
 
SV
Gertz
 
MA
Lacy
 
MQ
, et al. 
Thalidomide as initial therapy for early-stage myeloma.
Leukemia
2003
, vol. 
17
 (pg. 
775
-
779
)
31
Rajkumar
 
SV
Blood
 
E
Vesole
 
D
, et al. 
Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group.
J Clin Oncol
2006
, vol. 
24
 (pg. 
431
-
436
)
32
Cavo
 
M
Zamagni
 
E
Tosi
 
P
, et al. 
Superiority of thalidomide and dexamethasone over vincristine-doxorubicin-dexamethasone (VAD) as primary therapy in preparation for autologous transplantation for multiple myeloma.
Blood
2005
, vol. 
106
 (pg. 
35
-
39
)
33
Macro
 
M
Divine
 
M
Uzunhan
 
Y
, et al. 
Dexamethasone+thalidomide (dex/thal) compared to VAD as a pre-transplant treatment in newly diagnosed multiple myeloma (MM): a randomized trial [abstract].
Blood
2006
, vol. 
108
  
Abstract 57
34
Rajkumar
 
SV
Hayman
 
S
Gertz
 
MA
, et al. 
Combination therapy with thalidomide plus dexamethasone for newly diagnosed myeloma.
J Clin Oncol
2002
, vol. 
20
 (pg. 
3419
-
3423
)
35
Ludwig
 
H
Tothova
 
E
Hajek
 
R
, et al. 
Thalidomide-dexamethasone versus melphalan-prednisolone as first line treatment in elderly patients with multiple myeloma: second interim analysis.
Hematologica
2007
, vol. 
1
  
Abstract 446
36
Wang
 
M
Weber
 
D
Delasalle
 
K
Alexanian
 
R
Thalidomide–dexamethasone as primary therapy for advanced multiple myeloma.
Am J Hematol
2005
, vol. 
79
 (pg. 
194
-
197
)
37
Palumbo
 
A
Bringhen
 
S
Caravita
 
T
, et al. 
Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: randomised controlled trial.
Lancet
2006
, vol. 
367
 (pg. 
825
-
831
)
38
Facon
 
T
Mary
 
J
Hulin
 
C
, et al. 
Melphalan 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
2007
, vol. 
370
 (pg. 
1209
-
1218
)
39
Hulin
 
C
Facon
 
T
Rodon
 
P
, et al. 
Melphalan-Prednisone-Thalidomide (MP-T) demonstrates a significant survival advantage in elderly patients >75 years with multiple myeloma compared with Melphalan and Prednisone (MP) in a randomized, double blind, placebo-controlled trial IFM 01–01 [abstract].
Blood
2007
, vol. 
110
  
Abstract 75
40
Zervas
 
K
Dimopoulus
 
MA
Hatzicharissi
 
E
, et al. 
Primary treatment of multiple myeloma with thalidomide, vincristine, liposomal doxorubicin and dexamethasone (T-VAD doxil): a phase II multicenter study.
Ann Oncol
2004
, vol. 
15
 (pg. 
134
-
138
)
41
Osman
 
K
Comenzo
 
R
Rajkumar
 
SV
Deep venous thrombosis and thalidomide therapy for multiple myeloma.
N Engl J Med
2001
, vol. 
344
 pg. 
1951
 
42
Schutt
 
P
Ebeling
 
P
Buttkereit
 
U
, et al. 
Thalidomide in combination with vincristine, epirubicin and dexamethasone (VED) for previously untreated patients with multiple myeloma.
Eur J Haematol
2005
, vol. 
74
 (pg. 
40
-
46
)
43
Baz
 
R
Li
 
L
Kottke-Marchant
 
K
, et al. 
The role of aspirin in the prevention of thrombotic complications of thalidomide and anthracycline-based chemotherapy for multiple myeloma.
Mayo Clin Proc
2005
, vol. 
80
 (pg. 
1549
-
1551
)
44
Offidani
 
M
Corvatta
 
L
Piersantelli
 
MN
, et al. 
Thalidomide, dexamethasone and pegylated liposomal doxorubicin (ThaDD) for patients older than 65 years with newly diagnosed multiple myeloma.
Blood
2006
, vol. 
108
 (pg. 
2159
-
2164
)
45
Zervas
 
K
Mihou
 
D
Katodritou
 
E
, et al. 
VAD-doxil versus VAD-doxil plus thalidomide as initial treatment for multiple myeloma: results of a multicenter randomized trial of the Greek myeloma study group.
Ann Oncol
2007
, vol. 
18
 (pg. 
1369
-
1375
)
46
Minnema
 
MC
Breitkreutz
 
I
Auwerda
 
Jja
, et al. 
Prevention of venous thromboembolism with low molecular-weight heparin in patients with multiple myeloma treated with thalidomide and chemotherapy.
Leukemia
2004
, vol. 
18
 (pg. 
2044
-
2046
)
47
Wu
 
P
Davies
 
FE
Horton
 
C
, et al. 
The combination of cyclophosphomide, thalidomide and dexamethasone is an effective alternative to cyclophosphamide-vincristine-doxorubicin-methylprednisolone as induction chemotherapy prior to autologous transplantation for multiple myeloma: a case-matched analysis.
Leuk Lymphoma
2006
, vol. 
47
 (pg. 
2335
-
2338
)
48
Sidra
 
G
Williams
 
CD
Russel
 
NH
, et al. 
Combination chemotherapy with cyclophosphamide, thalidomide and dexamethasone for patients with refractory, newly diagnosed or relapsed myeloma.
Haematologica
2006
, vol. 
91
 (pg. 
862
-
863
)
49
Barlogie
 
B
Tricot
 
G
Anaissie
 
E
, et al. 
Thalidomide and hematopoietic-cell transplantation for multiple myeloma.
N Engl J Med
2006
, vol. 
354
 (pg. 
1021
-
1030
)
50
Zangari
 
M
Siegel
 
E
Barlogie
 
B
, et al. 
Thrombogenic activity of doxorubicin in myeloma patients receiving thalidomide: implications for therapy.
Blood
2002
, vol. 
100
 (pg. 
1168
-
1171
)
51
Zangari
 
M
Barlogie
 
B
Anaissie
 
E
, et al. 
Deep vein thrombosis in patients with multiple myeloma treated with thalidomide and chemotherapy: effects of prophylactic and therapeutic anticoagulation.
Br J Haematol
2004
, vol. 
126
 (pg. 
715
-
721
)
52
Rajkumar
 
SV
Hussein
 
M
Catalano
 
J
, et al. 
A multicenter, randomized, double-blind, placebo-controlled trial of thalidomide plus dexamethasone versus dexamethasone alone as initial therapy for newly diagnosed multiple myeloma [abstract].
J Clin Oncol
2006
, vol. 
24
 pg. 
7517
 
53
Breitkreutz
 
I
Lokhorst
 
HM
Raab
 
MS
, et al. 
Thalidomide in newly diagnosed multiple myeloma: influence of thalidomide treatment on peripheral blood stem cell collection yield.
Leukemia
2007
, vol. 
21
 (pg. 
1294
-
1299
)
54
Attal
 
M
Harousseau
 
JL
Leyvraz
 
S
, et al. 
Maintenance therapy with thalidomide improves survival in multiple myeloma patients.
Blood
2006
, vol. 
108
 (pg. 
3289
-
3294
)
55
Spencer
 
A
Prince
 
HM
Roberts
 
A
, et al. 
Thalidomide improve survivals when use after ASCT [abstract].
Hematologica
2007
, vol. 
92
  
Abstract S7b
56
Abdelkefi
 
A
Ladeb
 
S
Torjman
 
L
, et al. 
Single autologous stem cell transplantation followed by maintenance therapy with thalidomide is superior to double autologous transplantation in multiple myeloma: results of a multicentre randomized clinical trial.
Blood
2008
, vol. 
111
 (pg. 
1805
-
1810
)
57
Cavo
 
M
Testoni
 
N
Terragna
 
C
, et al. 
Up-front thalidomide-dexamethasone (thal) and double autologous transplantation (double TX) for multiple myeloma: comparison with double TX without added thalidomide and prognostic implications of chromosome 13 deletion and traslocation t(4;14) [abstract].
Blood
2006
, vol. 
108
  
Abstract 3081
58
Palumbo
 
A
Bringhen
 
S
Falco
 
P
, et al. 
Time to first progression, but not b2-microglobulin, predicts outcome in myeloma patients who received thalidomide as salvage therapy.
Cancer
2007
, vol. 
110
 (pg. 
824
-
829
)
59
Gunzler
 
V
Thalidomide in human immunodeficiency virus (HIV) patients: a review of safety considerations.
Drug Safety
1992
, vol. 
7
 (pg. 
116
-
134
)
60
Teo
 
S
Morgan
 
M
Stirling
 
D
, et al. 
Assessment of the in vitro and in vivo genotoxicity of thalomid (thalidomide).
Teratog Carcinog Mutagen
2000
, vol. 
20
 (pg. 
301
-
311
)
61
Smithells
 
D
Does thalidomide cause second generation birth defects?
Drug Safety
1998
, vol. 
19
 (pg. 
339
-
341
)
62
Zangari
 
M
Saghafifar
 
F
Anaissie
 
E
, et al. 
Activated protein C resistance in the absence of factor V Leiden mutation is a common finding in multiple myeloma and is associated with an increased risk of thrombotic complications.
Blood Coagul Fibrinolysis
2002
, vol. 
13
 (pg. 
187
-
192
)
63
Corso
 
A
Lorenzi
 
A
Terulla
 
V
, et al. 
Modification of thrombomodulin plasma levels in refractory myeloma patients during treatment with thalidomide and dexamethasone.
Ann Hematol
2004
, vol. 
83
 (pg. 
588
-
591
)
64
Kaushal
 
V
Kaushal
 
GP
Melkaveri
 
SN
, et al. 
Thalidomide protects endothelial cells from doxorubicin-induced apoptosis but alters cell morphology.
J Thromb Haemost
2004
, vol. 
2
 (pg. 
327
-
334
)
65
Streetly
 
M
Hunt
 
BJ
Parmar
 
K
, et al. 
Markers of endothelial and haemostatic function in the treatment of relapsed myeloma with the immunomodulatory agent actimid (CC-4047) and their relationship with venous thrombosis.
Eur J Haematol
2005
, vol. 
74
 (pg. 
293
-
296
)
66
Dimopoulos
 
MA
Eleutherakis
 
V
Adverse effects of thalidomide administration in patients with neoplastic disease.
Am J Med
2004
, vol. 
117
 (pg. 
508
-
515
)
67
Fahdi
 
IE
Gaddam
 
V
Saucedo
 
JF
, et al. 
Bradycardia during therapy for multiple myeloma with thalidomide.
Am J Cardiol
2004
, vol. 
93
 (pg. 
1052
-
1055
)
68
Mileshkin
 
L
Stark
 
R
Day
 
B
, et al. 
Development of neuropathy in patients with myeloma treated with thalidomide: patterns of occurrence and the role of electrophysiologic monitoring.
J Clin Oncol
2006
, vol. 
24
 (pg. 
4507
-
4514
)
69
Richardson
 
P
Schlossman
 
R
Jagannath
 
S
, et al. 
Thalidomide for patients with relapsed multiple myeloma after high-dose chemotherapy and stem cell transplantation: results of an open-label multicenter phase 2 study of efficacy, toxicity and biological activity.
Mayo Clin Proc
2004
, vol. 
79
 (pg. 
875
-
882
)
70
Cavaletti
 
G
Beronio
 
A
Reni
 
L
, et al. 
Thalidomide sensory neurotoxicity: a clinical and neurophysiologic study.
Neurology
2004
, vol. 
62
 pg. 
2291
 
71
Bastuji-Garin
 
S
Ochonisky
 
S
Bouche
 
P
, et al. 
Incidence and risk factors for thalidomide neuropathy: a prospective study of 135 dermatologic patients.
J Invest Dermatol
2002
, vol. 
119
 (pg. 
1020
-
1026
)
72
Rajkumar
 
SV
Gertz
 
MA
Witzig
 
TE
Life-threatening toxic epidermal necrolysis with thalidomide therapy for myeloma.
N Engl J Med
2000
, vol. 
343
 (pg. 
972
-
973
)
73
Hall
 
VC
El-Azhary
 
RA
Bouwhuis
 
S
, et al. 
Dermatologic side effects of thalidomide in patients with multiple myeloma.
J Am Acad Dermatol
2003
, vol. 
48
 (pg. 
548
-
552
)
74
Badros
 
AZ
Siegel
 
E
Bodenner
 
D
, et al. 
Hypothyroidism in patients with multiple myeloma following treatment with thalidomide.
Am J Med
2002
, vol. 
112
 (pg. 
412
-
413
)
75
Escudier
 
B
Lassau
 
N
Couanet
 
D
, et al. 
Phase II trial of thalidomide in renal cell carcinoma.
Ann Oncol
2002
, vol. 
13
 (pg. 
1029
-
1035
)
76
Trojan
 
A
Chasse
 
E
Gay
 
B
, et al. 
Severe hepatic toxicity due to thalidomide in relapsed multiple myeloma.
Ann Oncol
2003
, vol. 
14
 (pg. 
501
-
502
)
77
Eriksson
 
T
Hoglund
 
P
Turesson
 
I
, et al. 
Pharmacokinetics of thalidomide in patients with impaired renal function and while on and off dialysis.
J Pharm Pharmacol
2003
, vol. 
55
 (pg. 
1701
-
1706
)
78
Tosi
 
P
Zamagni
 
E
Cellini
 
C
, et al. 
Thalidomide alone or in combination with dexamethasone in patients with advanced, relapsed or refractory multiple myeloma and renal failure.
Eur J Haematol
2004
, vol. 
73
 (pg. 
98
-
103
)
Sign in via your Institution