The clinical outcomes of 169 patients enrolled in the first clinical trial of thalidomide for advanced or refractory myeloma are updated. Seventeen patients remain alive and 10 are event-free, with a median follow-up of 9.2 years. According to multivariate analysis of pretreatment variables, cytogenetic abnormalities, present in 47% of patients within 3 months of enrollment, and λ light chain isotype both affected overall survival and event-free survival adversely. Forty percent of the 58 patients lacking these 2 unfavorable features, one-half of whom had no disease recurrence, survived at least 6 years, in contrast to fewer than 5% among those with 1 or 2 risk features (P < .001). Patients who had received cumulative thalidomide doses in excess of 42 g in the first 3 months enjoyed superior overall and event-free survival. The poor outcome associated with λ-type myeloma may relate to its overrepresentation in molecularly defined high-risk disease gleaned from studies in newly diagnosed myeloma.

The prognosis for patients with multiple myeloma (MM) was radically improved when the melphalan dose-response effect could be safely exploited by applying autografts. Thus, the frequency of complete response (CR) was raised from less than 5% with standard melphalan-prednisone1  (MP) to 40% and higher when tandem transplants were applied.2  During the past 10 years, many new, mainly nongenotoxic, agents have been investigated. Thalidomide was the first since the introduction of melphalan and glucocorticoids to exhibit single-agent activity in MM that had become refractory to MP and even high-dose melphalan regimens.3,4  The recognition of thalidomide's activity in MM, long considered an “orphan disease” because of its relatively low incidence, ushered in an entirely new era of concerted clinical investigations of new agents, leading to US Food and Drug Administration approval of the first-in-class proteasome inhibitor bortezomib5  and the immunomodulatory agent lenalidomide,6  which are now being applied in the primary management of patients with MM.7-11 

Given the landmark observation of thalidomide's activity in end-stage MM, we now report on the long-term outcome of the 169 patients enrolled in the original phase 1/2 trial conducted at the University of Arkansas for Medical Sciences and in accordance with the Declaration of Helsinki.

We examined the long-term clinical outcomes of the original 169 patients who were enrolled on our UARK 98-003 protocol that called for administra-tion of thalidomide in a dose-escalated fashion for all patients according to tolerance, starting with 200 mg/d and allowing for dose increases every 2 weeks by 200 mg to a maximum dose of 800 mg/d.1,2  The protocol had been approved by the institutional review board of the University of Arkansas for Medical Sciences. All subjects had signed a written informed consent delineating the investigational nature of the trial, in line with institutional and Food and Drug Administration guidelines. We scrutinized the initial baseline characteristics and now provide follow-up information on event-free survival (EFS) and overall survival (OS) from start of protocol therapy, accounting for risk factors before therapy. Seventeen patients remain alive with a median follow-up of 9.2 years.

The presence of metaphase-based cytogenetic abnormalities (CAs) was identified in terms of CA subtypes, as previously published.12  Briefly, clinical outcomes were examined with regard to the presence of any CA and of the following subtypes: hypodiploidy/deletion 13, chromosome 1q amplification, chromosome 1p deletion, complete or partial deletion of chromosome 17, specific translocations, and the presence of myelodysplasia-associated abnormalities within a MM-typical CA.12 

Kaplan-Meier methods were used to generate survival distribution graphs, and comparisons were made using the log-rank test.13,14  Multivariate analyses of EFS and OS were done using stepwise selection and Cox proportional hazard regression modeling.15 

As of March 24, 2008 (median follow-up, 9.2 years), 10 patients remain event-free and 17 alive (Figure 1A). Twenty-five percent of patients remained event-free 2 years after discontinuing the study drug. The median time to next therapy was 3 months for the 53 patients receiving salvage regimens, which comprised further high-dose melphalan regimens in 29% and bortezomib in only 7%.

Figure 1

Survival outcomes after starting thalidomide therapy. (A) Overall and event-free survival for all 169 patients. At 6 years, 18% of the original 169 enrolled were alive and 8% event-free. (B) Overall and event-free survival according to cumulative thalidomide dose consumed within the first 3 months from protocol start. Patients whose cumulative thalidomide dose exceeded 42 g within 90 days of enrollment had significantly longer OS and EFS. (C) Overall and event-free survival comparisons according to the presence of cytogenetic abnormalities detected within 3 months before protocol enrollment. Patients without cytogenetic abnormalities (CA) enjoyed superior overall and event-free survival. (D) Overall and event-free survival according to the number of independent adverse parameters present before protocol enrollment (cytogenetic abnormalities (CA), λ light chain). Overall survival and event-free survival were superior in patients lacking cytogenetic abnormalities and exhibiting κ light chain isotypes.

Figure 1

Survival outcomes after starting thalidomide therapy. (A) Overall and event-free survival for all 169 patients. At 6 years, 18% of the original 169 enrolled were alive and 8% event-free. (B) Overall and event-free survival according to cumulative thalidomide dose consumed within the first 3 months from protocol start. Patients whose cumulative thalidomide dose exceeded 42 g within 90 days of enrollment had significantly longer OS and EFS. (C) Overall and event-free survival comparisons according to the presence of cytogenetic abnormalities detected within 3 months before protocol enrollment. Patients without cytogenetic abnormalities (CA) enjoyed superior overall and event-free survival. (D) Overall and event-free survival according to the number of independent adverse parameters present before protocol enrollment (cytogenetic abnormalities (CA), λ light chain). Overall survival and event-free survival were superior in patients lacking cytogenetic abnormalities and exhibiting κ light chain isotypes.

Close modal

Of the 140 patients with well-documented dosing information beyond 90 days of trial participation, 58% proceeded to the planned 800-mg target dose; 68% reached the 600-mg and 87% the 400-mg level, whereas 13% of patients did not proceed to doses beyond 200 mg. The median duration of maintaining the highest thalidomide dose level was 49 days for the 800-mg dose, 20 days for both 600-mg and 400-mg doses, and 13 days for the 200-mg dose level. As expected, those failing to proceed beyond the 400-mg dose level comprised significantly higher proportions of patients with high-risk features such as β2-microglobulin more than or equal to 3.5 mg/L (59% vs 31%, P = .002) and hypodiploidy (35% vs 11%, P = .026).

This phase 1/2 trial was not designed to define the optimal thalidomide dose but to enable maximum dose escalation, where feasible, in the absence of information on a thalidomide dose-response effect. Using a 90-day landmark, both OS and EFS were superior among subjects who had taken at least 42 g of study drug (median total dose; range, 0.5-963.5 g) during this time frame (Figure 1B).

The presence of CAs was associated with inferior 3-year OS and EFS rates of 14% and 1% compared with 52% and 22% in patients with normal karyotypes (Figure 1C). Previously not appreciated was the inferior outcome associated with λ versus κ light chain myeloma which, along with the presence of CAs, imparted inferior OS and EFS in multivariate analysis (Table 1). When specific CA subgroups were considered, deletion 13, along with renal insufficiency, dominated both OS and EFS models (Table 1). Among 141 patients with complete information, the 58 (41%) lacking CAs and presenting with κ-type myeloma constituted a favorable group whose 5-yr OS and EFS rates were 45% and 20%, respectively, compared with 5% and 2% for the remainder (Figure 1D).

Table 1

Univariate and multivariate analyses of variables associated with overall and event-free survival

Variablen/N (%)Overall survival
Event-free survival
HR (95% CI)PHR (95% CI)P
Univariate 
    IgA isotype* 43/154 (28) 1.17 (0.81-1.69) .408 1.21 (0.85-1.74) .293 
    λ light chain 46/153 (30) 1.48 (1.03-2.14) .036 1.43 (1.00-2.05) .048 
    Age ≥ 65 yr 43/169 (25) 1.55 (1.09-2.22) .016 1.44 (1.01-2.05) .045 
    CRP ≥ 4 mg/L 108/158 (68) 1.58 (1.09-2.28) .016 1.57 (1.09-2.24) .014 
    β2-microglobulin ≥ 3.5 mg/L 66/160 (41) 3.74 (2.62-5.34) < .001 1.89 (1.35-2.65) < .001 
    Albumin < 3.5 g/dL 35/163 (21) 1.91 (1.29-2.81) .001 1.18 (0.81-1.74) .386 
    Hb < 10 g/dL 67/163 (41) 2.28 (1.63-3.19) < .001 1.69 (1.22-2.36) .002 
    Platelets < 150 × 109/L 104/163 (64) 1.50 (1.06-2.11) .021 1.06 (0.76-1.47) .743 
    Creatinine ≥ 2.0 mg/dL 23/162 (14) 2.88 (1.82-4.57) < .001 2.11 (1.34-3.34) .001 
    LDH ≥ 190 U/L 39/160 (24) 1.81 (1.25-2.64) .002 1.42 (0.98-2.06) .061 
    ISS stage 1* 84/160 (53) 0.30 (0.21-0.42) < .001 0.55 (0.40-0.77) < .001 
    ISS stage 2* 37/160 (23) 1.85 (1.26-2.71) .002 1.38 (0.94-2.00) .098 
    ISS stage 3* 39/160 (24) 3.19 (2.16-4.71) < .001 1.68 (1.15-2.44) .007 
    Cytogenetic abnormalities (CAs) 75/161 (47) 3.07 (2.15-4.38) < .001 2.21 (1.57-3.12) < .001 
        Hypodiploid 24/149 (16) 2.49 (1.58-3.92) < .001 2.25 (1.42-3.54) < .001 
        Deletion 13q 38/154 (25) 2.76 (1.85-4.10) < .001 2.30 (1.56-3.39) < .001 
        Deletion 1p 11/154 (7) 2.20 (1.18-4.10) .014 2.03 (1.09-3.80) .026 
        Deletion 17p 11/154 (7) 1.93 (1.00-3.73) .049 1.94 (1.04-3.65) .039 
    At least 2 prior transplants 89/169 (53) 1.49 (1.07-2.06) .017 1.33 (0.97-1.82) .078 
Multivariate (any CA) 
    λ light chain 39/136 (29) 1.61 (1.08-2.42) .021 1.53 (1.04-2.25) .030 
    Age ≥ 65 years 31/136 (23) 2.03 (1.32-3.11) .001 NA NA 
    Cytogenetic abnormalities (CAs) 61/136 (45) 2.91 (1.95-4.35) < .001 2.23 (1.53-3.26) < .001 
    β2-microglobulin ≥ 3.5 mg/L 49/136 (36) 3.20 (2.15-4.76) < .001 NA NA 
Multivariate (CA subgroups) 
    Age ≥ 65 yr 30/130 (23) 1.92 (1.24-2.98) .003 NA NA 
    Creatinine ≥ 2.0 mg/dL 17/130 (13) 2.00 (1.09-3.70) .026 1.88 (1.10-3.21) .022 
    β2-microglobulin ≥ 3.5 mg/L 45/130 (35) 2.47 (1.55-3.92) < .001 NA NA 
    Deletion 13 28/130 (22) 2.18 (1.35-3.52) .002 2.17 (1.39-3.40) < .001 
    Hb < 10 g/dL 51/130 (39) 1.63 (1.08-2.47) .021 NA NA 
    At least 2 prior transplants 68/130 (52) 1.64 (1.11-2.42) .012 NA NA 
Variablen/N (%)Overall survival
Event-free survival
HR (95% CI)PHR (95% CI)P
Univariate 
    IgA isotype* 43/154 (28) 1.17 (0.81-1.69) .408 1.21 (0.85-1.74) .293 
    λ light chain 46/153 (30) 1.48 (1.03-2.14) .036 1.43 (1.00-2.05) .048 
    Age ≥ 65 yr 43/169 (25) 1.55 (1.09-2.22) .016 1.44 (1.01-2.05) .045 
    CRP ≥ 4 mg/L 108/158 (68) 1.58 (1.09-2.28) .016 1.57 (1.09-2.24) .014 
    β2-microglobulin ≥ 3.5 mg/L 66/160 (41) 3.74 (2.62-5.34) < .001 1.89 (1.35-2.65) < .001 
    Albumin < 3.5 g/dL 35/163 (21) 1.91 (1.29-2.81) .001 1.18 (0.81-1.74) .386 
    Hb < 10 g/dL 67/163 (41) 2.28 (1.63-3.19) < .001 1.69 (1.22-2.36) .002 
    Platelets < 150 × 109/L 104/163 (64) 1.50 (1.06-2.11) .021 1.06 (0.76-1.47) .743 
    Creatinine ≥ 2.0 mg/dL 23/162 (14) 2.88 (1.82-4.57) < .001 2.11 (1.34-3.34) .001 
    LDH ≥ 190 U/L 39/160 (24) 1.81 (1.25-2.64) .002 1.42 (0.98-2.06) .061 
    ISS stage 1* 84/160 (53) 0.30 (0.21-0.42) < .001 0.55 (0.40-0.77) < .001 
    ISS stage 2* 37/160 (23) 1.85 (1.26-2.71) .002 1.38 (0.94-2.00) .098 
    ISS stage 3* 39/160 (24) 3.19 (2.16-4.71) < .001 1.68 (1.15-2.44) .007 
    Cytogenetic abnormalities (CAs) 75/161 (47) 3.07 (2.15-4.38) < .001 2.21 (1.57-3.12) < .001 
        Hypodiploid 24/149 (16) 2.49 (1.58-3.92) < .001 2.25 (1.42-3.54) < .001 
        Deletion 13q 38/154 (25) 2.76 (1.85-4.10) < .001 2.30 (1.56-3.39) < .001 
        Deletion 1p 11/154 (7) 2.20 (1.18-4.10) .014 2.03 (1.09-3.80) .026 
        Deletion 17p 11/154 (7) 1.93 (1.00-3.73) .049 1.94 (1.04-3.65) .039 
    At least 2 prior transplants 89/169 (53) 1.49 (1.07-2.06) .017 1.33 (0.97-1.82) .078 
Multivariate (any CA) 
    λ light chain 39/136 (29) 1.61 (1.08-2.42) .021 1.53 (1.04-2.25) .030 
    Age ≥ 65 years 31/136 (23) 2.03 (1.32-3.11) .001 NA NA 
    Cytogenetic abnormalities (CAs) 61/136 (45) 2.91 (1.95-4.35) < .001 2.23 (1.53-3.26) < .001 
    β2-microglobulin ≥ 3.5 mg/L 49/136 (36) 3.20 (2.15-4.76) < .001 NA NA 
Multivariate (CA subgroups) 
    Age ≥ 65 yr 30/130 (23) 1.92 (1.24-2.98) .003 NA NA 
    Creatinine ≥ 2.0 mg/dL 17/130 (13) 2.00 (1.09-3.70) .026 1.88 (1.10-3.21) .022 
    β2-microglobulin ≥ 3.5 mg/L 45/130 (35) 2.47 (1.55-3.92) < .001 NA NA 
    Deletion 13 28/130 (22) 2.18 (1.35-3.52) .002 2.17 (1.39-3.40) < .001 
    Hb < 10 g/dL 51/130 (39) 1.63 (1.08-2.47) .021 NA NA 
    At least 2 prior transplants 68/130 (52) 1.64 (1.11-2.42) .012 NA NA 

NA indicates not applicable.

*

Variable not included for multivariate selection.

The listed toxicities have to be viewed with some caution because of possible underreporting by patients for fear of having to discontinue thalidomide, which was then the only available salvage option. The most common grade 3 or higher toxicities recorded included sensory/motor neuropathy and weakness in 20%, somnolence in 14%, and constipation in 12%; 7% experienced tremor and 5% complained of ataxia.

In this long-term follow-up of a quintessential trial of the first novel agent with antimyeloma efficacy in 3 decades, we note that 17 patients are still alive; 10 of the 17 have had no recurrence since trial initiation more than 9 years ago. Consistent with the initial report, the presence of CAs was the dominant feature linked to both inferior OS and EFS; deletion 13 was the only significant adverse genetic feature when individual CA subgroups were considered. A novel hitherto unrecognized favorable feature was the presence of a κ rather than λ light chain isotype. The poor outcome of patients with λ-type myeloma may be due to its association with high-risk molecular subtypes of myeloma.16  Gene expression profiling studies as part of Total Therapy 2 and 3 trials17,18  revealed an even distribution of κ and λ light chain myeloma (frequency ratio, 1.1) in 3 high-risk entities (MAF/MAFB, FGFR3/MMSET, and proliferation) compared with a strong κ light chain predominance (frequency ratio 2.3) in the remaining molecular subgroups with a superior prognosis (P < .001).

Thalidomide has since been effectively combined with dexamethasone,7  standard melphalan in MPT (melphalan, prednisone, thalidomide),9  and bortezomib in VTD (bortezomib, thalidomide, dexamethasone),11  resulting in remarkably high partial and complete response rates and promising survival duration. When used as part of a tandem transplant trial in Total Therapy 2, randomization to thalidomide resulted in superior CR rates and EFS17  and, according to a recent update, also in extension of OS, which was however limited to the CA subgroup,19  contradicting the findings reported in this salvage setting.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

This work was supported in part by the National Cancer Institute (P01 grant CA55819) and Celgene (Summit, NJ).

National Institutes of Health

Contribution: F.v.R., J.D.S., J.Z., and B.B. designed the study and wrote the manuscript; M.D. wrote the original protocol; F.v.R., E.A., D.S., S.S., J.M., S.J., and B.B. enrolled and cared for patients; B.J. collected data; and A.H. and J.C. analyzed data.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Bart Barlogie, Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205; e-mail: barlogiebart@uams.edu.

1
Alexanian
 
R
Haut
 
A
Khan
 
AU
, et al. 
Combination chemotherapy with different melphalan dose regimens.
JAMA
1969
, vol. 
208
 (pg. 
1680
-
1685
)
2
Barlogie
 
B
Jagannath
 
S
Vesole
 
DH
, et al. 
Superiority of tandem autologous transplantation over standard therapy for previously untreated multiple myeloma.
Blood
1997
, vol. 
89
 (pg. 
789
-
793
)
3
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
)
4
Barlogie
 
B
Desikan
 
R
Eddlemon
 
P
, et al. 
Extended 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
)
5
Richardson
 
PG
Barlogie
 
B
Berenson
 
J
, et al. 
A phase 2 study of bortezomib in relapsed, refractory myeloma.
N Engl J Med
2003
, vol. 
348
 (pg. 
2609
-
2617
)
6
Richardson
 
PG
Blood
 
E
Mitsiadis
 
CS
, et al. 
A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma.
Blood
2006
, vol. 
108
 (pg. 
3458
-
3464
)
7
Rajkumar
 
SV
Blood
 
E
Vesole
 
D
Fonseca
 
R
Greipp
 
PR
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 [abstract].
J Clin Oncol
2006
, vol. 
24
 (pg. 
431
-
436
)
8
Rajkumar
 
S
Jacobus
 
S
Callander
 
N
, et al. 
A randomized phase III trial of lenalidomide plus high-dose dexamethasone vs lenalidomide plus low-dose dexamethasone in newly diagnosed multiple myeloma (E4A03): a trial coordinated by the Eastern Cooperative Oncology Group [abstract].
Blood
2006
, vol. 
108
 pg. 
799
 
9
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: randomized controlled trial.
Lancet
2006
, vol. 
367
 (pg. 
825
-
831
)
10
Palumbo
 
A
Falco
 
P
Falcone
 
A
, et al. 
Oral revlimid plus melphalan and prednisone (R-MP) for newly diagnosed multiple myeloma: results of a multicenter phase I/II study [abstract].
Blood
2006
, vol. 
108
 pg. 
800a
 
11
Wang
 
M
Delassale
 
K
Giralt
 
S
, et al. 
Rapid control of previously untreated multiple myeloma with bortezomib-thalidomide-dexamethasone followed by early intensive therapy [abstract].
Blood
2005
, vol. 
106
 pg. 
784a
 
12
Jacobson
 
J
Barlogie
 
B
Shaughnessy
 
J
, et al. 
MDS-type abnormalities within myeloma signature karyotype (MM-MDS): only 13% 1-year survival despite tandem transplants.
Br J Haematol
2003
, vol. 
133
 (pg. 
430
-
440
)
13
Kaplan
 
EL
Meier
 
P
Nonparametric estimation from incomplete observations.
J Am Stat Assoc
1958
, vol. 
53
 (pg. 
457
-
458
)
14
Gooley
 
TA
Leisering
 
W
Crowley
 
J
Storer
 
BE
Estimation of failure probabilities in the presence of competing risks: new representations of old estimators.
Stat Med
1999
, vol. 
18
 (pg. 
695
-
706
)
15
Cox
 
DR
Regression tables and life tables.
J R Stat Soc B
1972
, vol. 
34
 (pg. 
187
-
202
)
16
Zhan
 
F
Huang
 
Y
Colla
 
S
, et al. 
The molecular classification of multiple myeloma.
Blood
2006
, vol. 
108
 (pg. 
2020
-
2028
)
17
Barlogie
 
B
Tricot
 
G
Anaissie
 
E
, et al. 
Thalidomide and hematopoietic-cell transplantation in multiple myeloma.
N Engl J Med
2006
, vol. 
354
 (pg. 
1021
-
1030
)
18
Pineda-Roma
 
M
Zangari
 
M
Haessler
 
J
, et al. 
Sustained complete remissions in multiple myeloma linked to bortezomib in total therapy 3: comparison with total therapy 2.
Br J Haematol
2007
, vol. 
140
 (pg. 
625
-
634
)
19
Barlogie
 
B
Shaughnessy
 
JD
Crowley
 
J
Superior survival among myeloma patients with cytogenetic abnormalities randomized to the thalidomide arm of total therapy 2.
N Engl J Med
2008
, vol. 
359
 (pg. 
210
-
212
)
Sign in via your Institution