Está en la página 1de 14

The new england journal of medicine

review article

drug therapy

Multiple Myeloma
Robert A. Kyle, M.D., and S. Vincent Rajkumar, M.D.

From the Division of Hematology and Inter-


nal Medicine, Mayo Clinic, Rochester, Min-
nesota. Address reprint requests to Dr. Kyle
at the Division of Hematology and Internal
Medicine, Mayo Clinic, 200 First St. S.W.,
Rochester, MN 55905, or at kyle.robert@
m
promising.2
ultiple myeloma is a plasma-cell neoplasm that is charac-
terized by skeletal destruction, renal failure, anemia, and hypercalcemia.1
Although myeloma remains incurable, recent advances in its treatment, in-
cluding the use of thalidomide and new drugs such as bortezomib and CC-5013, are
mayo.edu.

N Engl J Med 2004;351:1860-73. diagnosis


Copyright © 2004 Massachusetts Medical Society.

The most common symptoms on presentation are fatigue, bone pain, and recurrent in-
fections.3 New diagnostic criteria require the presence of at least 10 percent plasma
cells on examination of the bone marrow (or biopsy of a tissue with monoclonal plas-
ma cells), monoclonal protein in the serum or urine, and evidence of end-organ dam-
age.4,5 The end-organ damage that meets the criterion for the diagnosis consists of hy-
percalcemia, renal insufficiency, anemia, or bone lesions (a group of findings referred
to as CRAB).4 Occasional patients presenting without a detectable monoclonal protein
but otherwise meeting these diagnostic criteria are considered to have nonsecretory
myeloma. The serum free light-chain assay is useful for monitoring many patients with
oligosecretory and nonsecretory myeloma. The differential diagnosis includes mono-
clonal gammopathy of undetermined significance (MGUS), smoldering (asymptom-
atic) multiple myeloma, primary amyloidosis, solitary plasmacytoma, low-grade lym-
phoma, chronic lymphocytic leukemia, and metastatic carcinoma.4
The median length of survival after diagnosis is approximately three years. Recent
advances have identified new prognostic markers, such as the complete deletion of
chromosome 13 or its long arm, as detected by karyotyping; the t(4;14) or t(14;16)
translocation; and increased density of bone marrow microvessels. These complement
established markers of adverse outcomes, such as an increase in the plasma-cell–label-
ing index, increased levels of serum beta2-microglobulin, low levels of serum albumin,
plasmablastic features in the bone marrow, and circulating plasma cells.6

pathophysiology
evolution of mgus
The first pathogenetic step in the development of myeloma is the emergence of a lim-
ited number of clonal plasma cells, clinically known as MGUS.7 Patients with MGUS
do not have symptoms or evidence of end-organ damage, but they do have an annual
risk of 1 percent of progression to myeloma or a related malignant disease.7 Approx-
imately 50 percent of patients have translocations that involve the immunoglobulin
heavy-chain locus on chromosome 14q32 and one of five partner chromosomes,
11q13 (CCND1) (the most common), 4p16.3 (FGFR-3 and MMSET), 6p21 (CCND3),

1860 n engl j med 351;18 www.nejm.org october 28, 2004

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

16q23 (c-maf ), and 20q11 (mafB).8,9 These and of nuclear factor-kB (NF-kB) ligand (RANKL) and
other cytogenetic changes (Fig. 1) are thought to a reduction in the level of its decoy receptor, os-
play an important role in the evolution of MGUS. teoprotegerin.12 The increase in the ratio of RANKL
to osteoprotegerin results in the activation of os-
progression to myeloma teoclasts and bone resorption. Overexpression of
With progression of MGUS to myeloma, complex RANKL is probably mediated in part by the release
genetic events occur in the neoplastic plasma cell of macrophage inflammatory protein 1a by neo-
(Fig. 1).8 Changes also occur in the bone marrow plastic plasma cells.13 Improved understanding of
microenvironment, including the induction of myeloma-associated bone disease has led to the use
angiogenesis, the suppression of cell-mediated of prophylactic bisphosphonate therapy.9
immunity, and the development of paracrine sig-
naling loops involving cytokines such as interleu- therapy
kin-6 and vascular endothelial growth factor.10,11
The resultant interactions of myeloma cells, bone There is no evidence that early treatment of asymp-
marrow stromal cells, and microvessels contribute tomatic (smoldering) multiple myeloma is bene-
to persistence of the tumor and its resistance to ficial.14,15 The median time from diagnosis to the
drugs.9,10 Understanding the cellular microenvi- progression to symptomatic disease is two to three
ronment in myeloma has had a role in the develop- years. Two recent studies suggest that thalidomide
ment of drugs such as thalidomide and bortezomib, may delay the time to progression.16,17 However,
which are able to overcome chemotherapy-resis- because some patients may remain progression-
tant disease.10 free for several years, therapy for asymptomatic my-
The development of bone lesions in myeloma eloma is not recommended, given the toxic effects
is thought to be related to an increase in the ex- of thalidomide. An approach to the treatment of
pression by osteoblasts of the receptor activator newly diagnosed myeloma is illustrated in Figure 2.

Genomic instability
Microenvironmental changes
in bone marrow
Translocations
at 14q32
Increased
(50%)
angiogenesis

Deletion of
chromosome 13
(50%)

Myeloma
Increased bone
resorption
Normal cell

Infection?
Inflammation?
N-RAS, K-RAS (30%)
p16 Methylation (40%)
Secondary translocations?
Monoclonal gammopathy
of undetermined significance

Figure 1. Mechanisms of Disease Progression in the Monoclonal Gammopathies.


Percentages are the proportions of patients with the abnormality. Microenvironmental changes include increased bone resorption — medi-
ated by increased production of receptor activator of nuclear factor-kB ligand, decreased levels of osteoprotegerin, and increased levels of
macrophage inflammatory protein 1a; the induction of angiogenesis; increased levels of interleukin-6 and vascular endothelial growth factor;
and decreased immune surveillance.

n engl j med 351;18 www.nejm.org october 28, 2004 1861

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

dexamethasone for three to four months as induc-


Eligible for Not eligible for tion therapy.19 Despite an acceptable response rate,
transplantation transplantation this therapy and similar intravenous regimens have
disadvantages, including the need for an indwelling
central venous line and the risk of catheter-relat-
Induction Dexamethasone Melphalan plus prednisone ed infections, thrombotic events, and alopecia. Fur-
Therapy or or thermore, the role of doxorubicin and vincristine
vincristine, other alkylator-based therapy
doxorubicin until plateau is reached in the regimen consisting of vincristine, doxorubi-
and dexamethasone cin, and dexamethasone is limited, because dexa-
or
thalidomide–dexamethasone*
methasone alone contributes to most of the activi-
ty (Table 1).
An alternative choice for induction is the oral
Stem-cell harvest
regimen of thalidomide plus dexamethasone. In
a recent trial, 50 patients with newly diagnosed my-
eloma were treated with this combination.28 Tha-
lidomide (at a dose of 200 mg per day) was given
with dexamethasone (at a dose of 40 mg per day)
Consolidation Autologous Melphalan plus prednisone on days 1 through 4, 9 through 12, and 17 through
Therapy transplantation or
thalidomide–dexamethasone* 20 (odd cycles) and on days 1 through 4 (even cy-
until plateau is reached cles). Each cycle was 28 days long, and there was
typically no gap between the cycles unless time was
Second autologous needed for the resolution of toxic effects. The re-
transplantation* sponse rate in the patients in this trial was 64 per-
or
mini–allogeneic
cent, which is similar to that in previous trials with
transplantation* the regimen of vincristine, doxorubicin, and dexa-
methasone. No important problems were found in
the collection or engraftment of stem cells in the
patients after receiving this induction therapy.28,34
Deep-vein thrombosis was an unexpected adverse
Maintenance No treatment or corticosteroids* or thalidomide* event in 12 percent of the patients in this trial. In a
Therapy separate trial, the response rate with this regimen
was 72 percent, and the use of prophylactic antico-
agulation with warfarin or low-molecular-weight
Figure 2. An Approach to the Treatment of Newly Diagnosed Multiple Myeloma. heparin prevented the occurrence of deep-vein
Induction therapy for patients eligible for transplantation is administered thrombosis.17 In a randomized trial comparing tha-
in four cycles before stem cells are harvested. The term “plateau” indicates
a minimum of 12 months of therapy and the achievement of stable disease
lidomide plus dexamethasone with dexamethasone
for at least 2 months. Asterisks denote investigational therapies. alone, the response rate with thalidomide plus dex-
amethasone was significantly better (P=0.01).35

induction therapy in patients not eligible


When possible, patients should be encouraged to for transplantation
participate in clinical trials at the time of diagnosis Patients who are not eligible for transplantation
and beyond. because of age, poor physical condition, or coexist-
ing conditions receive standard therapy with alkyl-
induction therapy in patients eligible ating agents.36 Although vincristine, doxorubicin,
for autologous stem-cell transplantation and dexamethasone, dexamethasone alone, or tha-
Patients who are eligible for autologous stem-cell lidomide plus dexamethasone can also be used as
transplantation are first treated with a regimen that initial therapy for these patients, the oral regimen
is not toxic to hematopoietic stem cells. The use of of melphalan plus prednisone is preferable in this
alkylating agents is best avoided, because they can setting to minimize toxic effects, unless there is a
prevent an adequate mobilization of stem cells.18 need for a rapid response, such as in patients with
Many physicians use vincristine, doxorubicin, and large, painful lytic lesions or with worsening renal

1862 n engl j med 351;18 www.nejm.org october 28 , 2004

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

Table 1. Major Classes of Drugs Used in the Treatment of Myeloma.*


Drug Regimen and Usual Starting Dose Response Rate Trial
Newly
Diagnosed Relapsed
Disease Disease
percentage†
Alkylating agents
Melphalan and prednisone‡ Repeated every 6 wk 50–55 — Myeloma Trialists’ Collaborative
Melphalan, 8–10 mg PO on days 1–7 Group20
Prednisone, 60 mg per day PO on days 1–7
Combinations, e.g., VBMCP§ Repeated every 5 wk 60 15¶ Myeloma Trialists’ Collaborative
Vincristine, 0.03 mg/kg (maximum 2 mg) IV Group,20 Cavo et al.21
on day 1
Carmustine, 0.5 mg/kg IV on day 1
Melphalan, 0.25 mg/kg PO on days 1–7
Cyclophosphamide, 10 mg/kg IV on day 1
Prednisone, 1 mg/kg PO on days 1–7
Corticosteroids
Pulsed dexamethasone Repeated every 4–5 wk 45 25–35 Alexanian et al.,22 Alexanian et
Dexamethasone, 40 mg PO on days 1–4, al.23
9–12, and 17–20
VAD Repeated every 4 wk 55–65 25–50 Alexanian et al.,19 Alexanian et
Vincristine, 0.4 mg per day IV continuous al.,22 Alexanian et al.24
infusion on days 1–4
Doxorubicin, 9 mg/m2 IV continuous infu-
sion on days 1–4
Dexamethasone, 40 mg PO on days
1–4, 9–12, and 17–20
Thalidomide and its analogues
Thalidomide Repeated every 4 wk 35 25–45 Rajkumar et al.,16 Weber et al.,17
200–400 mg PO on days 1–28 Singhal et al.,25 Kumar et al.,26
Juliusson et al.27
Thalidomide–dexamethasone Repeated every 4 wk 65–70 50 Weber et al.,17 Rajkumar et al.,28
Thalidomide, 200 mg PO on days 1–28 Dimopoulos et al.29
Dexamethasone, 40 mg PO on days 1–4,
9–12, and 17–20¿
Melphalan–prednisone– Repeated every 4 wk 80 — Palumbo et al.30
thalidomide Melphalan, 4 mg/m2 PO on days 1–7
Prednisone, 40 mg/m2 PO on days 1–7
Thalidomide, 100 mg PO on days 1–28
Cyclophosphamide–thalido- Repeated every 3 wk — 75 Garcia-Sanz et al.31
mide–dexamethasone Cyclophosphamide, 50 mg PO on days 1–21
Thalidomide, 200–800 mg PO on days 1–21
Dexamethasone, 40 mg PO on days 1–4
CC-5013** 25–30 mg PO on days 1–21 every 28 days — 25 Richardson et al.32
Bortezomib 1.3 mg/m2 on days 1, 4, 8, and 11 every 21 — 25 Richardson et al.33
days

* PO denotes orally; VBMCP vincristine, carmustine, melphalan, cyclophosphamide, and prednisone; VAD vincristine, doxorubicin, and dex-
amethasone; IV intravenous; mg/kg milligrams per kilogram of body weight; and mg/m2 milligrams per square meter of body-surface area.
† Percentages are estimates based on the results of all studies in which the given regimen was used.
‡ Appropriate trials of melphalan–prednisone in patients with relapsed or refractory disease who have not received alkylator therapy have not
been conducted.
§ Several drug combinations have been studied; VBMCP is the combination most commonly studied and used.
¶ The response rate reflects effectiveness in patients with relapsed or refractory disease and previous alkylator therapy.
¿ In one study, dexamethasone was administered on days 1–4, 9–12, and 17–20 in odd cycles and on days 1–4 in even cycles.
** CC-5013 is not commercially available.

n engl j med 351;18 www.nejm.org october 28, 2004 1863

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

function. Despite better response rates with any of Generally, issues such as the inconvenience and
the more aggressive combination regimens than the adverse effects of prolonged chemotherapy and
with melphalan plus prednisone (Table 1), no sur- difficulties in gaining the approval of health insur-
vival benefit has been shown.20 Melphalan is gen- ance plans for cryopreservation of stem cells still
erally administered at a dose of 8 to 10 mg per day favor early autologous stem-cell transplantation.
for 7 days (although lower doses may be needed in This is especially the case for patients younger than
patients with advanced renal failure) with predni- 65 years of age with adequate renal function.46
sone at a dose of 60 mg per day orally during the
same 7 days, and both drugs are repeated every tandem transplantation
6 weeks for a period of 12 to 18 months.36 The dos- In tandem (double) autologous stem-cell trans-
age of melphalan is adjusted to produce mild cy- plantation, patients undergo a second planned au-
topenia at midcycle. tologous stem-cell transplantation after they have
recovered from the first. Tandem transplantation
autologous stem-cell transplantation was developed by Barlogie and colleagues, to im-
Although not curative, autologous stem-cell trans- prove complete-response rates.53,54 In a recent ran-
plantation improves the likelihood of a complete domized trial conducted in France, event-free sur-
response, prolongs disease-free survival and over- vival and overall survival were significantly better
all survival, and is a major advance in myeloma ther- among recipients of tandem transplantation than
apy (Table 2).46,47 The mortality rate is 1 to 2 per- among those who underwent a single autologous
cent, and approximately 50 percent of patients can stem-cell transplantation (P=0.01)43 (Table 2). Con-
be treated entirely as outpatients. Whether or not versely, preliminary data from three other random-
a complete response is achieved is an important ized trials showed no convincing improvement in
predictor of the eventual outcome. Melphalan (at overall survival among patients receiving tandem
a dose of 200 mg per square meter of body-surface transplantation, although the follow-up was too
area) is the most widely used preparative regimen short for definite conclusions to be drawn44,45,55
for autologous stem-cell transplantation and is su- (Table 2). On the basis of the results of the French
perior to the older regimen of melphalan (140 mg trial, it is reasonable to consider tandem transplan-
per square meter) combined with 8 Gy of total- tation for patients who do not have at least a very
body irradiation.48 The data are limited on the ef- good partial response (defined as a reduction of
fectiveness of autologous stem-cell transplanta- 90 percent or more in monoclonal protein levels)
tion in patients 65 years of age or older and those with the first transplantation.43 However, until this
with end-stage renal disease. However, the proce- issue is resolved, it may be advantageous to collect
dure is feasible in these patients and can be under- enough stem cells to allow a patient to undergo two
taken after careful consideration of the possible transplantations, reserving a second autologous
risks and benefits, perhaps with the use of an in- stem-cell transplantation for relapse.56
termediate dose of melphalan (100 mg per square Two challenges persist. First, the current con-
meter).49 ditioning regimens are inadequate. In an effort to
The role of autologous stem-cell transplanta- improve them, the use of bone-seeking radioac-
tion in the treatment of patients responding to in- tive holmium and samarium is being tested.57,58
duction therapy has been questioned. In a Spanish Second, reinfused hematopoietic stem cells are in-
trial, patients responding to initial therapy had a evitably contaminated with tumor cells. Such con-
similar overall survival and progression-free surviv- tamination can be reduced with the use of CD34
al with either autologous stem-cell transplantation selection or the delivery of in vitro chemotherapy
or eight additional courses of chemotherapy 39 — to the collected stem cells. However, no effect on
suggesting that patients who benefit most from overall survival has been observed to date, reflect-
autologous stem-cell transplantation are those with ing the fact that residual drug-resistant cells in the
disease refractory to induction therapy.50,51 The val- marrow are the principal cause of the recurrence
ue of autologous stem-cell transplantation as ini- of disease.59,60
tial therapy has also been challenged by the results
of three randomized trials that showed that trans- allogeneic transplantation
plantation can be safely delayed and used as salvage The advantages of allogeneic transplantation are
therapy at the time of relapse (Table 2).52 a graft that is not contaminated with tumor cells

1864 n engl j med 351;18 www.nejm.org october 28 , 2004

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
Table 2. Results of Major Randomized Trials with Autologous Stem-Cell Transplantation in Myeloma.

No. of
Trial Comparison Patients Outcome Comments Study
Intergroupe Francophone Conventional-dose chemotherapy vs. 200 Superior event-free survival and overall All patients <65 yr of age, all received Attal et al.37
du Myélome 90 autologous bone marrow trans- survival with transplantation; 5-yr interferon maintenance therapy,
plantation survival, 52 percent with transplan- and survival with chemotherapy
tation vs. 12 percent with chemo- was shorter than expected
therapy (P=0.03)
Medical Research Council Conventional-dose chemotherapy vs. 401 Superior progression-free survival and All patients <65 yr of age Child et al.38
Myeloma VII autologous stem-cell transplan- overall survival with transplanta-
tation tion; median survival, 54 mo with
transplantation vs. 42 mo with che-
motherapy (P=0.04)
PETHEMA* Conventional-dose chemotherapy vs. 216 No difference in progression-free sur- Median age, 56 yr; only 164 patients re- Bladé et al.39
autologous stem-cell transplan- vival or overall survival; median sponding to induction chemother-

n engl j med 351;18


tation survival, 65 mo with transplanta- apy underwent randomization
tion vs. 67 mo with chemotherapy
Myélome Autogreffe 91 Conventional-dose chemotherapy vs. 190 No significant difference in event-free All patients 55–65 yr of age Fermand et al.40
autologous stem-cell transplan- survival or overall survival; median
tation survival, 55 mo with transplanta-
tion vs. 50 mo with chemotherapy

www.nejm.org
drug therapy

Intergroup S9321 Conventional-dose chemotherapy vs. 549 Median progression-free survival, 25 After 39 patients were enrolled, alloge- Barlogie et al.41
autologous stem-cell transplan- mo with autologous transplanta- neic transplantation group closed;
tation vs. allogeneic transplantation tion vs. 21 mo with chemotherapy 52 percent of patients assigned to
(P=0.05); no significant difference chemotherapy received salvage
in overall survival transplantation at relapse
Myélome Autogreffe Early vs. delayed autologous stem-cell 185 Median event-free survival, 39 mo with All patients <56 yr of age; early trans- Fermand et al.42
transplantation autologous transplantation vs. 13 plantation associated with shorter

october 28, 2004


mo with chemotherapy; no signifi- duration of chemotherapy and
cant difference in overall survival symptoms
Intergroupe Francophone du Single vs. double autologous stem-cell 399 Superior event-free survival and overall All patients <60 yr of age; benefit of Attal et al.43
Myélome 94 transplantation survival with double transplantation; second transplantation restricted
7-yr survival, 42 percent with double to those with less than very good

Copyright © 2004 Massachusetts Medical Society. All rights reserved.


transplantation vs. 21 percent with partial response to first
single transplantation (P=0.01)
Bologna 96 Single vs. double autologous stem-cell 220 Superior event-free survival with dou- Interim analysis of first 220 patients Cavo et al.44
transplantation ble transplantation; no significant
difference in overall survival; medi-
an survival, 60 mo with double
transplantation vs. 56 mo with sin-
gle transplantation

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Myélome Autogreffe 95 Single vs. double autologous stem-cell 230 No difference in progression-free or All patients <56 yr of age Fermand et al.45
transplantation overall survival

* PETHEMA denotes Programa para el Estudio y Tratamiento de las Hemopatías Malignas.

1865
The new england journal of medicine

and a graft-versus-myeloma effect.61,62 However, ed that maintenance with prednisone may be use-
only 5 to 10 percent of patients are candidates for ful after conventional chemotherapy. The progres-
allogeneic transplantation when age, the availabil- sion-free survival was significantly longer with
ity of an HLA-matched sibling donor, and adequate 50 mg of oral, alternate-day prednisone (for a pe-
organ function are taken into consideration. Fur- riod of 14 months) than with 10 mg (for a period
thermore, the high rate of treatment-related death of 5 months; P=0.003). The overall survival was
has made conventional allogeneic transplanta- better with the use of the higher dose of predni-
tion unacceptable for most patients with myelo- sone, as compared with the lower dose (37 months
ma. T-cell–depleted transplants appear to be inef- and 26 months, respectively; P=0.05). It is unclear
fective.63 whether these results can be generalized, because
Several recent trials have used nonmyeloabla- this comparison study included only patients whose
tive conditioning regimens (also referred to as myeloma was responsive to corticosteroids and who
“mini” allogeneic transplantation).64 The greatest had not previously undergone autologous stem-cell
benefit has been reported in patients with newly di- transplantation. Clinical trials are under way to
agnosed disease who have first undergone autolo- evaluate novel approaches, such as the use of tha-
gous stem-cell transplantation to reduce the tumor lidomide and dendritic-cell vaccination as main-
burden and afterward undergone mini–allogeneic tenance therapy.
(nonmyeloablative) transplantation of stem cells
from an HLA-identical sibling donor.65 The rate of therapy for relapsed and refractory
treatment-related deaths was 15 to 20 percent with multiple myeloma
this strategy.66 There is also a high risk of both Almost all patients with multiple myeloma have
acute and chronic graft-versus-host disease, al- a risk of eventual relapse. If relapse occurs more than
though the emergence of these toxic effects ap- six months after conventional therapy is stopped,
pears to be necessary for disease control. Prelimi- the initial chemotherapy regimen should be re-
nary results from a French trial indicated that in instituted. Patients who have had stem cells cryopre-
patients with high-risk myeloma (those with a de- served early in the course of the disease can benefit
letion of chromosome 13 plus high levels of beta2- from the use of autologous stem-cell transplanta-
microglobulin), the overall survival with this ap- tion as salvage therapy.70
proach may not be superior to that with tandem The highest response rates in relapsed myelo-
autologous stem-cell transplantation.67 ma have been with the use of intravenous vincris-
Currently, the use of autologous stem-cell trans- tine, doxorubicin, and dexamethasone (Table 1).
plantation followed by mini–allogeneic transplan- Intravenous doxorubicin hydrochloride liposome
tation remains investigational and is best performed is a less cardiotoxic alternative to doxorubicin and
as part of a clinical trial. Outside that setting, we is being tested in patients with newly diagnosed
consider this option only for selected high-risk pa- disease.71 Dexamethasone alone is also effective.
tients with newly diagnosed disease who are young- Intravenous pulsed methylprednisolone (at a dose
er than 60 years of age and have HLA-identical sib- of 2 g three times per week) is an alternative to dex-
lings as potential donors, and for whom the current amethasone and may have fewer adverse effects.72
approaches appear to be futile (i.e., patients with In the past five years, major advances have been
karyotypic deletion of chromosome 13 or hypo- made with the use of thalidomide and the arrival of
diploidy, the t(4;14) or t(14;16) translocation, or novel approaches such as bortezomib. Depend-
a plasma cell–labeling index of 3 percent or more). ing on the clinical situation, these and other agents
(Table 1) are generally used sequentially, because
maintenance therapy disease refractory to one regimen or agent may re-
Initial trials of the usefulness of maintenance ther- spond to another.
apy with interferon alfa produced conflicting re-
sults, and the results of a meta-analysis showed thalidomide
only a modest improvement in overall survival.68
Recent results from an intergroup trial showed no Thalidomide was used as a sedative in the 1950s
apparent benefit from the use of interferon as main- and was withdrawn from the market after initial re-
tenance therapy.41 ports of teratogenicity in 1961.73 Subsequently, the
A study by Berenson and colleagues69 indicat- efficacy of thalidomide in erythema nodosum lep-

1866 n engl j med 351;18 www.nejm.org october 28 , 2004

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

rosum, Behçet’s syndrome, the wasting and oral ul- about 25 percent in patients receiving the drug in
cers associated with the human immunodeficiency combination with other cytotoxic chemotherapeu-
virus syndrome, and graft-versus-host disease per- tic agents, particularly doxorubicin.77,79,86-88 Other
mitted its use in clinical trials and for compassion- adverse effects include edema, bradycardia, neutro-
ate use.74 In 1998, the Food and Drug Administra- penia, impotence, and hypothyroidism. The use of
tion (FDA) approved the use of thalidomide in the thalidomide in pregnancy is absolutely contrain-
treatment of erythema nodosum leprosum. dicated, and the System for Thalidomide Educa-
tion and Prescribing Safety Program89 must be fol-
clinical trials lowed to prevent teratogenic effects.
Trials with thalidomide as an anticancer agent
were unsuccessful in the 1960s.75,76 However, the dosage
finding of increased angiogenesis in myeloma cou- Thalidomide is usually administered in a dosage of
pled with the recognition of the antiangiogenic 200 mg per day, which is increased to 400 mg per
properties of thalidomide led to the first clinical day after two to four weeks, if tolerated. Lower dos-
trial of this drug for the treatment of multiple my- es (50 to 100 mg) are being investigated, and to min-
eloma, at the University of Arkansas. In that trial, imize long-term toxic effects, the dose should be
the rate of response was 25 percent in patients with adjusted to the lowest level that can achieve and
relapsed and refractory disease.25 Since then, sev- maintain a response. Doses above 200 mg are gen-
eral studies have confirmed the activity of thalid- erally not indicated when thalidomide is used in
omide in relapsed myeloma, with response rates combination with corticosteroids or chemotherapy.
ranging from 25 percent to 35 percent.77-79 The re-
sponses are durable, with a median duration of ap- mechanism of action
proximately 12 months.26,77 Thalidomide had lim- Thalidomide undergoes rapid interconversion be-
ited activity in extramedullary (soft-tissue) disease tween the R-enantiomer and the S-enantiomer and
in one study.80 spontaneous cleavage to more than 12 metabolites
Given the activity of thalidomide as a single in solutions at physiologic pH.90 Furthermore, its
agent, subsequent trials explored its use in com- activity in most in vitro assays is moderate or negli-
bination with other active agents in the treatment gible, and its effects in animal models are depen-
of relapsed myeloma. Response rates when tha- dent on the species and the route of administra-
lidomide was used with corticosteroids, as com- tion. Thus, the study of its mechanism of action is
pared with the rates with thalidomide alone, in- difficult.91 Proposed mechanisms include the in-
creased to approximately 50 percent29,81-83 and to hibition of tumor necrosis factor a, the preven-
more than 70 percent when used in a three-drug tion of free-radical–mediated DNA damage, the
combination of thalidomide, dexamethasone, and suppression of angiogenesis, an increase in cell-
an alkylating agent (either cyclophosphamide or mediated cytotoxic effects, and the alteration of the
melphalan).31,79,84 Thalidomide alone or in com- expression of cellular adhesion molecules.74 Tha-
bination is now considered standard therapy for lidomide may also inhibit the activity of NF-kB
relapsed and refractory myeloma. As discussed ear- and the enzymes cyclooxygenase-1 and cyclooxy-
lier, this activity also has translated into the incor- genase-2 (Fig. 3).
poration of thalidomide in the initial treatment
of multiple myeloma. bortezomib
adverse effects Bortezomib (formerly known as PS-341) was the
Sedation, fatigue, constipation, and rash are com- first proteasome inhibitor to enter clinical trials. It
mon adverse effects but usually are responsive to was granted accelerated approval by the FDA for
dose reduction.85 Peripheral neuropathy occurs with the treatment of advanced myeloma in May 2003.
long-term use and often necessitates the discon-
tinuation of the therapy or a dose reduction. The clinical trials
incidence of deep-vein thrombosis is only 1 to 3 per- In preclinical models, bortezomib showed sub-
cent in patients receiving thalidomide alone but in- stantial activity against many cancers, including
creases to 10 to 15 percent in patients receiving the myeloma.92-96 Its promising efficacy against my-
drug in combination with dexamethasone and to eloma was noted in a phase 1 dose-finding study

n engl j med 351;18 www.nejm.org october 28, 2004 1867

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

had relapsed after the initial induction therapy and


Stromal cells consolidation therapy also was completed recent-
Thalidomide
ly.101 In this trial, patients were randomly assigned
Bortezomib to receive one of two doses of bortezomib (28 pa-
Thalidomide tients were assigned to receive a dose of 1.0 mg per
Bortezomib square meter and 26 patients 1.3 mg per square
Tumor necrosis factor a
Interleukin-6
meter) administered on days 1, 4, 8, and 11 in a
Alkylating agents 21-day cycle for a total of eight cycles. Responses
Corticosteroids Nuclear occurred in 33 percent of those receiving 1.0 mg
Bortezomib factor-kB Bortezomib
per square meter and in 50 percent of those receiv-
ing 1.3 mg per square meter. Although initial trials
Thalidomide allowed a maximum of only eight cycles of bortez-
omib, recent data indicate that it is safe to admin-
T cells ister at least an additional five or six cycles of ther-
apy without undue toxic effects.101 A recent phase
3 trial involving 670 patients and comparing bor-
Thalidomide
Natural killer cells tezomib with pulsed dexamethasone therapy was
closed early because of a longer time to disease pro-
gression in patients receiving bortezomib.102 In
Figure 3. Proposed Mechanism of Action of Drugs to Target the Myeloma Cell trials to date, the response to bortezomib has been
and Components of the Bone Marrow Microenvironment.
shown to be rapid, usually occurring within one or
In myeloma cells, alkylating agents, corticosteroids, and bortezomib inhibit
two cycles of the therapy.
cell growth and induce apoptosis. The effect of bortezomib on myeloma cells
is mediated in part by the inhibition of nuclear factor-kB. Thalidomide and Studies are under way of bortezomib in com-
bortezomib inhibit the interaction between myeloma cells and stromal cells bination with other effective agents. In the initial
as well as the production of cytokines such as tumor necrosis factor a and phase 2 trial, dexamethasone was added to therapy
interleukin-6. Thalidomide inhibits angiogenesis and stimulates the immune- for 106 patients whose condition had not respond-
surveillance properties of T cells and natural killer cells. Solid arrows indicate
ed or those in whom the disease had progressed
stimulation or secretion and dashed arrows inhibition.
while they were receiving bortezomib.33,103 In 19
of these patients (18 percent), there was a response
to the addition of dexamethasone with a reduction
of at least 25 percent in monoclonal protein levels,
conducted by Orlowski and colleagues.97 On the suggesting an additive effect that can be exploit-
basis of these observations, a phase 2 multicenter ed in future trials. Other investigators are studying
trial of intravenous bortezomib in myeloma was bortezomib in combination with thalidomide, peg-
initiated33 (the drug was given at a dose of 1.3 mg ylated doxorubicin, and alkylating agents.104,105
per square meter over a period of three to five sec-
onds on days 1, 4, 8, and 11 in a 21-day cycle for a adverse effects
maximum of eight cycles). Of 193 patients who The most common adverse effects of bortezomib
could be evaluated, 92 percent had received three are gastrointestinal symptoms, cytopenia, fatigue,
or more of the major classes of agents for myelo- and peripheral neuropathy.33,106 A decrease in the
ma, and in 91 percent the disease had been re- platelet count to less than 50,000 per cubic milli-
fractory to the most recent treatment. The partial- meter occurs in almost 30 percent of patients. Pe-
response rate with bortezomib was 27 percent, ripheral neuropathy, often painful, develops in ap-
and 4 percent of patients achieved a complete re- proximately 30 percent of patients and is more
sponse.98 The median duration of response was frequent in those who have previously received neu-
12 months, and the responses were associated with rotoxic therapy and those with a preexisting neu-
improvement in cytopenia, renal function, and the ropathy.
quality of life.99 Older age (above 65 years) and ex-
tensive marrow involvement were associated with dosing
a lower rate of response.33,100 The recommended starting dose is 1.3 mg per
A randomized, phase 2 trial of bortezomib in square meter administered on days 1, 4, 8, and 11
myeloma that had not responded to treatment or of a 21-day cycle.33 Reductions to 1.0 mg per square

1868 n engl j med 351;18 www.nejm.org october 28, 2004

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

meter or, if needed, to 0.7 mg per square meter may duction of at least 50 percent in monoclonal pro-
be necessary, depending on the toxic effects. tein levels in approximately 30 percent of patients
with relapsed myeloma, and myelosuppression was
mechanism of action the major dose-limiting toxic effect.114,115 A multi-
Bortezomib is a specific inhibitor of the 26S pro- center, randomized phase 2 study in which the sub-
teasome, a large intracellular adenosine triphos- jects had relapsed refractory myeloma was recently
phate–dependent protease responsible for protein completed,32 in which CC-5013 was administered
catabolism in all eukaryotic cells. Normally, cellular at a dose of 15 mg twice per day or a dose of 30 mg
proteins destined for catabolism are first ubiquit- once per day for a period of 3 weeks and was fol-
inated — a pathway in which C-terminal glycine res- lowed by a 1-week rest (in a 28-day cycle). The cycle
idues of ubiquitin molecules attach covalently to was repeated until disease progression or toxic ef-
specific lysine moieties on the protein.107 Ubiquit- fects occurred. In 24 percent of 83 patients who
inated proteins are identified and degraded in the could be evaluated, there was a reduction of at least
central portion of the proteasome, a pathway criti- 50 percent in monoclonal protein levels. The most
cal for normal cellular events to occur, including cell common adverse effects were grade 3 or higher
cycling, signal transduction, and transcriptional thrombocytopenia (i.e., a platelet count of less than
regulation. Inhibition of this pathway creates ma- 50,000 per cubic millimeter), which occurred in
jor imbalances in the levels of various regulatory 18 percent of patients, and neutropenia (i.e., a neu-
proteins, leading to arrest of the cell cycle and apo- trophil count of less than 1000 per cubic milli-
ptosis. meter), which occurred in 28 percent of patients.
The therapeutic effect of bortezomib-induced Adverse events were more frequent among those
inhibition of the proteasome in myeloma is prob- receiving 15 mg twice per day. Common adverse
ably a result of direct cytotoxicity and of effects on effects observed with thalidomide, however, such
the bone marrow microenvironment (Fig. 3).94,108 as sedation, constipation, and neuropathy, were not
One of the consequences of proteasome inhibi- observed. CC-5013 appears to be a promising agent
tion is the accumulation of IkB, an inhibitor of the for the treatment of myeloma, and trials conducted
major transcription factor NF-kB.109 At the cellu- for approval by the FDA are under way.
lar level, inhibition of NF-kB leads to a decrease
in the expression of adhesion molecules and vari- other novel agents
ous growth, survival, and angiogenic factors. It also
causes a decrease in the levels of the proteins that Several other agents, including 2-methoxyestra-
promote apoptosis, Bcl-2 and A1/Bfl-1, triggering diol, neovastat, oblimersen, farnesyltransferase,
the release of cytochrome-c, activation of caspase and histone deacetylase inhibitors, are being ac-
9, and apoptosis of myeloma cells.94 However, in- tively investigated.10,112 Preliminary evidence sug-
hibition of NF-kB is unlikely to be the sole mecha- gests that arsenic trioxide has clinical activity against
nism of the observed antimyeloma effects.92,94,110 myeloma,116 and trials are under way to confirm
this possibility and to determine optimal dosing.
cc-5013 Another thalidomide analogue, CC-4047, also has
shown activity.117
As a means to overcoming the nonhematologic tox-
ic effects of thalidomide, including teratogenicity, treatment of complications
several active analogues of thalidomide have been
developed. CC-5013 (lenalidomide) is an amino- Table 3 summarizes current management strate-
substituted variant of thalidomide that belongs to gies for complications observed in patients with my-
a class of analogues known as immunomodulatory eloma. Important advances include the prevention
drugs. Its preclinical activity is more potent and and treatment of hypercalcemia and bony lesions
more promising than the activity of thalidomide. with the monthly administration of bisphospho-
The drug induces apoptosis and decreases the bind- nates in patients with myeloma bone disease.118,119
ing of myeloma cells to stromal cells in bone mar- Such treatment may be complicated by albumin-
row.111 It also inhibits angiogenesis and promotes uria, renal dysfunction, and osteonecrosis of the
cytotoxicity mediated by natural killer cells.112,113 jaw. Another recent advance is the use of vertebro-
In two phase 2 trials of CC-5013, there was a re- plasty or kyphoplasty to reduce pain and help re-

n engl j med 351;18 www.nejm.org october 28, 2004 1869

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

Table 3. Treatment of Complications in Multiple Myeloma.

Complication Therapeutic Options


Myeloma bone disease Pamidronate or zoledronic acid in patients with documented bone disease
Encouragement of activity to prevent osteopenia and deep-vein thrombosis
Pain control with narcotic analgesics, if needed; avoidance of nonsteroidal antiinflam-
matory agents
Radiation to treat painful bony lesions refractory to pain medication or cord compression
Surgical intervention to prevent or treat pathologic fractures
Vertebroplasty or kyphoplasty for selected vertebral lesions, to reduce pain and improve
height
Anemia Treatment of reversible causes such as deficiencies of iron, B12, or folate
Erythropoietin for symptomatic anemia during chemotherapy
Transfusions as needed
Infections Vaccination against Streptococcus pneumoniae, Haemophilus influenzae, and influenza
Consideration of prophylactic broad-spectrum antibiotic therapy when corticosteroids
are used
Intravenous immune globulin for recurrent serious infections associated with hypogam-
maglobulinemia
Consideration of prophylaxis against Pneumocystis carinii when prolonged corticosteroid
therapy is used; avoidance of trimethoprim–sulfamethoxazole when thalidomide
is used
Hypercalcemia Intravenous fluids and corticosteroids
Bisphosphonates when hypercalcemia is severe or unresponsive to hydration and corti-
costeroids
Renal failure Correction of reversible causes such as dehydration, hypercalcemia, and hyperuricemia
Chemotherapy (e.g., vincristine, doxorubicin, and dexamethasone; dexamethasone
alone; or thalidomide–dexamethasone) for rapid control of disease
Alkaline diuresis for acute renal failure due to cast nephropathy; avoid alkalinization in
patients with hypercalcemia
Trial of plasma exchange in acute evolving renal failure
Hyperviscosity syndrome Plasma exchange for symptomatic patients (serum viscosity does not correlate well with
symptoms)

store vertebral height in patients with compression neously, one to improve rates of complete response
fractures. with the intense use of available methods, which is
aimed at finding a cure, and the other to exploit
future directions conventional and novel agents in an effort to con-
trol myeloma and convert it into a chronic indolent
Ongoing randomized trials promise to define fur- disease. Success in either direction would be mo-
ther the roles of new agents, mini–allogeneic trans- mentous.
plantation, and maintenance therapy. There is a Supported in part by grants from the National Cancer Institute
continuing search for active agents based on ad- (CA62242, CA85818, CA93842, CA107476, and CA100080), the Mul-
tiple Myeloma Research Foundation, and the Leukemia and Lym-
vances in understanding the biology of myelo- phoma Society.
ma.112 Microarray-based pharmacogenomic analy- Dr. Kyle reports having received consulting fees from NeoRx,
sis may usher in an era of therapy tailored to the Millennium, Aeterna, Celgene, and Novartis; and Dr. Rajkumar
consultation fees from Celgene and grants from Celgene and Mil-
individual patient. 120
lennium.
Finally, two paths are being explored simulta-

references
1. Kyle RA, Rajkumar SV. Plasma cell dis- 3. Kyle RA, Gertz MA, Witzig TE, et al. Re- related disorders: a report of the Interna-
orders. In: Goldman L, Ausiello DA, eds. view of 1027 patients with newly diagnosed tional Myeloma Working Group. Br J Hae-
Cecil textbook of medicine. 22nd ed. Phila- multiple myeloma. Mayo Clin Proc 2003; matol 2003;121:749-57.
delphia: W.B. Saunders, 2004:1184-95. 78:21-33. 5. Rajkumar SV, Dispenzieri A, Fonseca
2. Anderson KC. Multiple myeloma: how 4. International Myeloma Working Group. R, et al. Thalidomide for previously untreated
far have we come? Mayo Clin Proc 2003;78: Criteria for the classification of monoclo- indolent or smoldering multiple myeloma.
15-7. nal gammopathies, multiple myeloma and Leukemia 2001;15:1274-6.

1870 n engl j med 351;18 www.nejm.org october 28, 2004

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

6. Rajkumar SV, Greipp PR. Prognostic 22. Alexanian R, Dimopoulos MA, Dela- multiple myeloma (E1A00): a trial coordi-
factors in multiple myeloma. Hematol Oncol salle K, Barlogie B. Primary dexamethasone nated by the Eastern Cooperative Oncology
Clin North Am 1999;13:1295-314. treatment of multiple myeloma. Blood 1992; Group. Prog Proc Am Soc Clin Oncol 2004;
7. Kyle RA, Therneau TM, Rajkumar SV, 80:887-90. 23:558. abstract.
et al. A long-term study of prognosis in 23. Alexanian R, Barlogie B, Dixon D. 36. Rajkumar SV, Gertz MA, Kyle RA,
monoclonal gammopathy of undetermined High-dose glucocorticoid treatment of resis- Greipp PR. Current therapy for multiple my-
significance. N Engl J Med 2002;346:564-9. tant myeloma. Ann Intern Med 1986;105: eloma. Mayo Clin Proc 2002;77:813-22.
8. Kuehl WM, Bergsagel PL. Multiple my- 8-11. 37. Attal M, Harousseau JL, Stoppa AM, et
eloma: evolving genetic events and host in- 24. Alexanian R, Barlogie B, Ventura G. al. A prospective, randomized trial of autol-
teractions. Nat Rev Cancer 2002;2:175-87. Chemotherapy for resistant and relapsing ogous bone marrow transplantation and
9. Seidl S, Kaufmann H, Drach J. New in- multiple myeloma. Eur J Haematol Suppl chemotherapy in multiple myeloma. N Engl
sights into the pathophysiology of multiple 1989;51:140-4. J Med 1996;335:91-7.
myeloma. Lancet Oncol 2003;4:557-64. 25. Singhal S, Mehta J, Desikan R, et al. 38. Child JA, Morgan GJ, Davies FE, et al.
10. Hideshima T, Anderson KC. Molecular Antitumor activity of thalidomide in refrac- High-dose chemotherapy with hematopoi-
mechanisms of novel therapeutic approaches tory multiple myeloma. N Engl J Med 1999; etic stem-cell rescue for multiple myeloma.
for multiple myeloma. Nat Rev Cancer 2002; 341:1565-71. [Erratum, N Engl J Med 2000; N Engl J Med 2003;348:1875-83.
2:927-37. 342:364.] 39. Bladé J, Sureda A, Ribera JM, et al. High-
11. Rajkumar SV, Mesa RA, Fonseca R, et 26. Kumar S, Gertz MA, Dispenzieri A, et dose therapy autotransplantation/intensifi-
al. Bone marrow angiogenesis in 400 patients al. Response rate, durability of response, cation versus continued conventional che-
with monoclonal gammopathy of undeter- and survival after thalidomide therapy for motherapy in multiple myeloma patients
mined significance, multiple myeloma, and relapsed multiple myeloma. Mayo Clin Proc responding to initial chemotherapy: defin-
primary amyloidosis. Clin Cancer Res 2002; 2003;78:34-9. itive results from PETHEMA after a median
8:2210-6. 27. Juliusson G, Celsing F, Turesson I, Len- follow-up of 66 months. Blood 2003;102:
12. Roodman GD. Role of the bone marrow hoff S, Adriansson M, Malm C. Frequent 42a-43a. abstract.
microenvironment in multiple myeloma. good partial remissions from thalidomide 40. Fermand J-P, Ravaud P, Katsahian S, et
J Bone Miner Res 2002;17:1921-5. including best response ever in patients with al. High dose therapy (HDT) and autolo-
13. Abe M, Hiura K, Wilde J, et al. Role for advanced refractory and relapsed myeloma. gous blood stem cell (ABSC) transplantation
macrophage inflammatory protein (MIP)-1a Br J Haematol 2000;109:89-96. versus conventional treatment in multiple
and MIP-1b in the development of osteolytic 28. Rajkumar SV, Hayman S, Gertz MA, myeloma (MM): results of a randomized trial
lesions in multiple myeloma. Blood 2002; et al. Combination therapy with thalido- in 190 patients 55 to 65 years of age. Blood
100:2195-202. mide plus dexamethasone for newly diag- 1999;94:Suppl 1:396a. abstract.
14. Hjorth M, Hellquist L, Holmberg E, nosed myeloma. J Clin Oncol 2002;20:4319- 41. Barlogie B, Kyle R, Anderson K, et al.
Magnusson B, Rödjer S, Westin J. Initial ver- 23. Comparable survival in multiple myeloma
sus deferred melphalan-prednisone therapy 29. Dimopoulos MA, Zervas K, Kouvatseas (MM) with high dose therapy (HDT) employ-
for asymptomatic multiple myeloma stage I G, et al. Thalidomide and dexamethasone ing MEL 140 mg/m2 + TBI 12 Gy autotrans-
— a randomized study. Eur J Haematol combination for refractory multiple myelo- plants versus standard dose therapy with
1993;50:95-102. ma. Ann Oncol 2001;12:991-5. VBMCP and no benefit from interferon (IFN)
15. Grignani G, Gobbi PG, Formisano R, 30. Palumbo A, Bertola A, Musto P, et al. maintenance: results of intergroup trial
et al. A prognostic index for multiple my- Oral melphalan, prednisone and thalidomide S9321. Blood 2003;102:42a. abstract.
eloma. Br J Cancer 1996;73:1101-7. for newly diagnosed myeloma. Blood 2003; 42. Fermand JP, Ravaud P, Chevret S, et al.
16. Rajkumar SV, Gertz MA, Lacy MQ, et al. 102:148a. abstract. High-dose therapy and autologous periph-
Thalidomide as initial therapy for early- 31. Garcia-Sanz R, Gonzalez-Fraile MI, eral blood stem cell transplantation in mul-
stage myeloma. Leukemia 2003;17:775-9. Sierra M, Lopez C, Gonzalez M, San tiple myeloma: up-front or rescue treatment?
17. Weber D, Rankin K, Gavino M, Dela- Miguel JF. The combination of thalido- Results of a multicenter sequential random-
salle K, Alexanian R. Thalidomide alone or mide, cyclophosphamide and dexametha- ized clinical trial. Blood 1998;92:3131-6.
with dexamethasone for previously untreated sone (ThaCyDex) is feasible and can be an 43. Attal M, Harousseau JL, Facon T, et al.
multiple myeloma. J Clin Oncol 2003;21: option for relapsed/refractory multiple my- Single versus double autologous stem-cell
16-9. eloma. Hematol J 2002;3:43-8. transplantation for multiple myeloma. N Engl
18. Goldschmidt H, Hegenbart U, Wall- 32. Richardson PG, Jagannath S, Schloss- J Med 2003;349:2495-502. [Erratum, N Engl
meier M, Hohaus S, Haas R. Factors influenc- man R, et al. A multi-center, randomized, J Med 2004;350:2628.]
ing collection of peripheral blood progenitor phase 2 study to evaluate the efficacy and 44. Cavo M, Tosi P, Zamagni E, et al. The
cells following high-dose cyclophospha- safety of 2 CDC-5013 dose regimens when “Bologna 96” clinical trial of single vs. dou-
mide and granulocyte colony-stimulating used alone or in combination with dexa- ble autotransplants for previously untreated
factor in patients with multiple myeloma. Br methasone (Dex) for the treatment of relapsed multiple myeloma patients. Blood 2002;100:
J Haematol 1997;98:736-44. or refractory multiple myeloma (MM). Blood 179a. abstract.
19. Alexanian R, Barlogie B, Tucker S. VAD- 2003;102:235a. abstract. 45. Fermand J-P, Alberti C, Marolleau J-P.
based regimens as primary treatment for 33. Richardson PG, Barlogie B, Berenson J, Single versus tandem high dose therapy
multiple myeloma. Am J Hematol 1990;33: et al. A phase 2 study of bortezomib in re- (HDT) supported with autologous blood
86-9. lapsed, refractory myeloma. N Engl J Med stem cell (ABSC) transplantation using unse-
20. Myeloma Trialists’ Collaborative Group. 2003;348:2609-17. lected or CD34-enriched ABSC: results of a
Combination chemotherapy versus mel- 34. Ghobrial IM, Dispenzieri A, Bundy KL, two by two designed randomized trial in 230
phalan plus prednisone as treatment for et al. Effect of thalidomide on stem cell col- young patients with multiple myeloma (MM).
multiple myeloma: an overview of 6,633 lection and engraftment in patients with Hematol J 2003;4:Suppl 1:S59. abstract.
patients from 27 randomized trials. J Clin multiple myeloma. Bone Marrow Transplant 46. Bladé J, Vesole DH, Gertz M. Trans-
Oncol 1998;16:3832-42. 2003;32:587-92. plantation for multiple myeloma: who, when,
21. Cavo M, Galieni P, Tassi C, Gobbi M, 35. Rajkumar SV, Blood E, Vesole DH, how often? Patient selection and goals. Blood
Tura S. M-2 protocol for melphalan-resis- Shepard R, Greipp PR. A randomised phase 2003;102:3469-77.
tant and relapsing multiple myeloma. Eur III trial of thalidomide plus dexamethasone 47. Kumar A, Loughran T, Alsina M, Durie
J Haematol 1988;40:168-73. versus dexamethasone in newly diagnosed BG, Djulbegovic B. Management of multiple

n engl j med 351;18 www.nejm.org october 28, 2004 1871

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

myeloma: a systematic review and critical ized controlled trial. J Clin Oncol 2001;19: 72. Gertz MA, Garton JP, Greipp PR, Wit-
appraisal of published studies. Lancet Oncol 3771-9. zig TE, Kyle RA. A phase II study of high-
2003;4:293-304. 60. Barbui AM, Galli M, Dotti G, et al. Neg- dose methylprednisolone in refactory or re-
48. Moreau P, Facon T, Attal M, et al. Com- ative selection of peripheral blood stem cells lapsed multiple myeloma. Leukemia 1995;
parison of 200 mg/m2 melphalan and 8 Gy to support a tandem autologous transplan- 9:2115-8.
total body irradiation plus 140 mg/m2 mel- tation programme in multiple myeloma. Br J 73. Lenz W. Thalidomide and congenital
phalan as conditioning regimens for periph- Haematol 2002;116:202-10. abnormalities. Lancet 1962;1:45.
eral blood stem cell transplantation in 61. Mehta J, Singhal S. Graft-versus- 74. Rajkumar SV, Mesa RA, Tefferi A.
patients with newly diagnosed multiple myeloma. Bone Marrow Transplant 1998;22: A review of angiogenesis and anti-angio-
myeloma: final analysis of the Intergroupe 835-43. genic therapy in hematologic malignancies.
Francophone du Myélome 9502 randomized 62. Gahrton G, Svensson H, Cavo M, et al. J Hematother Stem Cell Res 2002;11:33-47.
trial. Blood 2002;99:731-5. Progress in allogeneic bone marrow and 75. Grabstald H, Golbey R. Clinical experi-
49. Palumbo A, Bringhen S, Petrucci MT, et peripheral blood stem cell transplantation ences with thalidomide in patients with
al. A prospective randomized trial of inter- for multiple myeloma: a comparison between cancer. Clin Pharmacol Ther 1965;40:298-
mediate dose melphalan (100 mg/m2) vs. transplants performed 1983-93 and 1994-98 302.
oral melphalan/prednisone. Blood 2003;102: at European Group for Blood and Marrow 76. Olson KB, Hall TC, Horton J, Khung CL,
984a. abstract. Transplantation centres. Br J Haematol 2001; Hosley HF. Thalidomide (N-phthaloylgluta-
50. Rajkumar SV, Fonseca R, Lacy MQ, et 113:209-16. mimide) in the treatment of advanced can-
al. Autologous stem cell transplantation for 63. Lokhorst HM, Segeren CM, Verdonck cer. Clin Pharmacol Ther 1965;6:292-7.
relapsed and primary refractory myeloma. LF, et al. Partially T-cell–depleted allogeneic 77. Barlogie B, Desikan R, Eddlemon P, et
Bone Marrow Transplant 1999;23:1267-72. stem-cell transplantation for first-line treat- al. Extended survival in advanced and refrac-
51. Bladé J, Esteve J. Treatment approaches ment of multiple myeloma: a prospective tory multiple myeloma after single-agent tha-
for relapsing and refractory multiple my- evaluation of patients treated in the phase III lidomide: identification of prognostic fac-
eloma. Acta Oncol 2000;39:843-7. study HOVON 24 MM. J Clin Oncol 2003;21: tors in a phase 2 study of 169 patients. Blood
52. Facon T, Mary JY, Harousseau JL, et al. 1728-33. 2001;98:492-4.
Front-line or rescue autologous bone mar- 64. Einsele H, Schafer HJ, Hebart H, et al. 78. Rajkumar SV, Fonseca R, Dispenzieri A,
row transplantation (ABMT) following a Follow-up of patients with progressive mul- et al. Thalidomide in the treatment of re-
first course of high dose melphalan (HDM) tiple myeloma undergoing allografts after lapsed multiple myeloma. Mayo Clin Proc
in multiple myeloma (MM): preliminary re- reduced-intensity conditioning. Br J Haema- 2000;75:897-901.
sults of a prospective randomized trial tol 2003;121:411-8. 79. Dimopoulos MA, Anagnostopoulos A,
(CIAM protocol). Blood 1996;88:Suppl 1: 65. Maloney DG, Sahebi F, Stockerl-Gold- Weber D. Treatment of plasma cell dyscra-
685a. abstract. stein KE, et al. Combining an allogeneic sias with thalidomide and its derivatives.
53. Barlogie B, Jagannath S, Vesole DH, et graft-vs-myeloma effect with high-dose J Clin Oncol 2003;21:4444-54.
al. Superiority of tandem autologous trans- autologous stem cell rescue in the treatment 80. Bladé J, Perales M, Rosinol L, et al. Tha-
plantation over standard therapy for previ- of multiple myeloma. Blood 2001;98:434a- lidomide in multiple myeloma: lack of re-
ously untreated multiple myeloma. Blood 435a. abstract. sponse of soft-tissue plasmacytomas. Br J
1997;89:789-93. 66. Maloney DG, Molina AJ, Sahebi F, et al. Haematol 2001;113:422-4.
54. Barlogie B, Jagannath S, Desikan KR, et Allografting with nonmyeloablative condi- 81. Weber DM, Rankin K, Gavino M, Dela-
al. Total therapy with tandem transplants tioning following cytoreductive autografts salle K, Alexanian R. Thalidomide with dex-
for newly diagnosed multiple myeloma. for the treatment of patients with multiple amethasone for resistant multiple myeloma.
Blood 1999;93:55-65. myeloma. Blood 2003;102:3447-54. Blood 2000;96:167a. abstract.
55. Goldschmidt H. Single vs. tandem au- 67. Moreau P, Garban F, Facon T, et al. Pre- 82. Palumbo A, Giaccone L, Bertola A, et al.
tologous transplantation in multiple my- liminary results of the IFM9903 and Low-dose thalidomide plus dexamethasone
eloma: the GMMG experience. Hematol J IFM9904 protocols comparing autologous is an effective salvage therapy for advanced
2003;4:Suppl 1:S61. abstract. followed by miniallogeneic transplantation myeloma. Haematologica 2001;86:399-403.
56. Sirohi B, Powles R, Singhal S, et al. and double autologous transplant in high- 83. Anagnostopoulos A, Weber D, Rankin
High-dose melphalan and second autografts risk de novo multiple myeloma. Blood 2003; K, Delasalle K, Alexanian R. Thalidomide
for myeloma relapsing after one autograft: 102:43a. abstract. and dexamethasone for resistant multiple
results equivalent to tandem autotransplan- 68. Myeloma Trialists’ Collaborative Group. myeloma. Br J Haematol 2003;121:768-71.
tation. Blood 2001;98:402a. abstract. Interferon as therapy for multiple myeloma: 84. Srkalovic G, Elson P, Trebisky B, Karam
57. Bensinger W, Giralt S, Holmberg L, et an individual patient data overview of 24 ran- MA, Hussein MA. Use of melphalan, thalid-
al. 166H0-DOTMP and high-dose melphalan domized trials and 4012 patients. Br J Hae- omide, and dexamethasone in treatment of
before autologous peripheral blood stem matol 2001;113:1020-34. refractory and relapsed multiple myeloma.
cell transplantation in patients with multi- 69. Berenson JR, Crowley JJ, Grogan TM, et Med Oncol 2002;19:219-26.
ple myeloma. Hematol J 2003;4:Suppl 1: al. Maintenance therapy with alternate-day 85. Ghobrial IM, Rajkumar SV. Manage-
S215-S216. abstract. prednisone improves survival in multiple ment of thalidomide toxicity. J Supportive
58. Dispenzieri A, Wiseman GA, Lacy MQ, myeloma patients. Blood 2002;99:3163-8. Oncol 2003;1:194-205.
et al. A phase II study of high dose 153-sa- 70. Gertz MA, Lacy MQ, Inwards DJ, et al. 86. Osman K, Comenzo R, Rajkumar SV.
marium EDTMP (153-Sm EDTMP) and mel- Early harvest and late transplantation as an Deep venous thrombosis and thalidomide
phalan for peripheral stem cell transplanta- effective therapeutic strategy in multiple therapy for multiple myeloma. N Engl J Med
tion (PBSCT) in multiple myeloma (MM). myeloma. Bone Marrow Transplant 1999;23: 2001;344:1951-2.
Blood 2003;102:982a. abstract. 221-6. 87. Zangari M, Anaissie E, Barlogie B, et al.
59. Stewart AK, Vescio R, Schiller G, et al. 71. Hussein MA, Wood L, Hsi E, et al. Increased risk of deep-vein thrombosis in
Purging of autologous peripheral-blood A phase II trial of pegylated liposomal doxo- patients with multiple myeloma receiving
stem cells using CD34 selection does not rubicin, vincristine, and reduced-dose dexa- thalidomide and chemotherapy. Blood 2001;
improve overall or progression-free survival methasone combination therapy in newly 98:1614-5.
after high-dose chemotherapy for multiple diagnosed multiple myeloma patients. Can- 88. Zangari M, Siegel E, Barlogie B, et al.
myeloma: results of a multicenter random- cer 2002;95:2160-8. Thrombogenic activity of doxorubicin in

1872 n engl j med 351;18 www.nejm.org october 28, 2004

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

myeloma patients receiving thalidomide: 100. Richardson PGG, Barlogie B, Beren- tiple myeloma. J Biol Chem 2002;277:16639-
implications for therapy. Blood 2002;100: son JR, et al. Prognostic factors associated 47.
1168-71. with response in patients with relapsed and 111. Mitsiades N, Mitsiades CS, Poulaki V,
89. Zeldis JB, Williams BA, Thomas SD, El- refractory multiple myeloma (MM) treated et al. Apoptotic signaling induced by immu-
sayed ME. S.T.E.P.S.: a comprehensive pro- with bortezomib. Prog Proc Am Soc Clin nomodulatory thalidomide analogs in human
gram for controlling and monitoring access Oncol 2003;22:581. abstract. multiple myeloma cells: therapeutic implica-
to thalidomide. Clin Ther 1999;21:319-30. 101. Berenson JR, Jagannath S, Barlogie B, tions. Blood 2002;99:4525-30.
90. Eriksson T, Bjorkman S, Hoglund P. et al. Experience with long-term therapy 112. Hideshima T, Chauhan D, Podar K,
Clinical pharmacology of thalidomide. Eur using the proteasome inhibitor, bortezo- Schlossman RL, Richardson P, Anderson KC.
J Clin Pharmacol 2001;57:365-76. mib, in advanced multiple myeloma (MM). Novel therapies targeting the myeloma cell
91. Bauer KS, Dixon SC, Figg WD. Inhibi- Prog Proc Am Soc Clin Oncol 2003;22:581. and its bone marrow microenvironment.
tion of angiogenesis by thalidomide requires abstract. Semin Oncol 2001;28:607-12.
metabolic activation, which is species-depen- 102. Richardson P, Sonneveld P, Schuster 113. Dredge K, Marriott JB, Macdonald CD,
dent. Biochem Pharmacol 1998;55:1827-34. MW, et al. Bortezomib vs. dexamethasone in et al. Novel thalidomide analogues display
92. Hideshima T, Richardson P, Chauhan relapsed multiple myeloma: a phase 3 ran- anti-angiogenic activity independently of
D, et al. The proteasome inhibitor PS-341 domized study. Prog Proc Am Soc Clin Oncol immunomodulatory effects. Br J Cancer
inhibits growth, induces apoptosis, and over- 2004;23:558. abstract. 2002;87:1166-72.
comes drug resistance in human multiple 103. Jagannath S, Richardson P, Barlogie B, 114. Zangari M, Tricot G, Zeldis J, Eddlemon
myeloma cells. Cancer Res 2001;61:3071-6. et al. SUMMIT/CREST Investigators. Phase P, Saghafifar F, Barlogie B. Results of phase I
93. Adams J, Palombella VJ, Sausville EA, et II trials of bortezomib in combination study of CC-5013 for the treatment of multi-
al. Proteasome inhibitors: a novel class of with dexamethasone in multiple myeloma ple myeloma (MM) patients who relapse af-
potent and effective antitumor agents. Can- (MM): assessment of additional benefits to ter high dose chemotherapy (HDCT). Blood
cer Res 1999;59:2615-22. combination in patients with sub-optimal 2001;98:775a. abstract.
94. Mitsiades N, Mitsiades CS, Poulaki V, et responses to bortezomib alone. Prog Proc 115. Richardson PG, Schlossman RL, Weller
al. Molecular sequelae of proteasome inhibi- Am Soc Clin Oncol 2003;22:582. abstract. E, et al. Immunomodulatory drug CC-5013
tion in human multiple myeloma cells. 104. Yang HH, Vescio RA, Adams J, Schen- overcomes drug resistance and is well tol-
Proc Natl Acad Sci U S A 2002;99:14374-9. kein D, Berenson JR. A phase I/II study of erated in patients with relapsed multiple
95. Mitsiades N, Mitsiades CS, Richardson combination treatment with bortezomib myeloma. Blood 2002;100:3063-7.
PG, et al. The proteasome inhibitor PS-341 and melphalan (Vc+M) in patients with 116. Munshi NC. Arsenic trioxide: an
potentiates sensitivity of multiple myeloma relapsed or refractory multiple myeloma emerging therapy for multiple myeloma.
cells to conventional chemotherapeutic (MM). Prog Proc Am Soc Clin Oncol 2003; Oncologist 2001;6:Suppl 2:17-21.
agents: therapeutic applications. Blood 22:582. abstract. 117. Streetly M, Jones RW, Knight R, et al.
2003;101:2377-80. 105. Orlowski RZ, Voorhees PM, Garcia R, An update of the use and outcomes of the
96. LeBlanc R, Catley LP, Hideshima T, et al. et al. Phase I study of the proteasome inhib- new immunomodulatory agent CC-4047
Proteasome inhibitor PS-341 inhibits hu- itor bortezomib and pegylated liposomal (actimid) in patients with relapsed/refractory
man myeloma cell growth in vivo and pro- doxorubicin in patients with refractory hema- myeloma. Blood 2003;102:236a. abstract.
longs survival in a murine model. Cancer tologic malignancies. Prog Proc Am Soc Clin 118. Berenson JR, Lichtenstein A, Porter L,
Res 2002;62:4996-5000. Oncol 2003;22:200. abstract. et al. Efficacy of pamidronate in reducing
97. Orlowski RZ, Stinchcombe TE, Mitchell 106. Richardson P. Proteasome inhibitors skeletal events in patients with advanced
BS, et al. Phase I trial of the proteasome in- in hematologic malignancies. Cancer Treat multiple myeloma. N Engl J Med 1996;334:
hibitor PS-341 in patients with refractory Rev 2003;29:Suppl 1:33-9. 488-93.
hematologic malignancies. J Clin Oncol 107. Almond JB, Cohen GM. The protea- 119. Berenson JR, Rosen LS, Howell A, et
2002;20:4420-7. some: a novel target for cancer chemother- al. Zoledronic acid reduces skeletal-related
98. Bladé J, Samson D, Reece D, et al. Crite- apy. Leukemia 2002;16:433-43. events in patients with osteolytic metasta-
ria for evaluating disease response and pro- 108. Hideshima T, Mitsiades C, Akiyama M, ses. Cancer 2001;91:1191-200. [Erratum,
gression in patients with multiple myeloma et al. Molecular mechanisms mediating anti- Cancer 2001;91:1956.]
treated by high-dose therapy and haemopoi- myeloma activity of proteasome inhibitor 120. Shaughnessy J Jr. Primer on medical
etic stem cell transplantation. Br J Haematol PS-341. Blood 2003;101:1530-4. genomics. IX. Scientific and clinical appli-
1998;102:1115-23. 109. Palombella VJ, Rando OJ, Goldberg cations of DNA microarrays — multiple
99. Lee S, Richardson PG, Barlogie B, et al. AL, Maniatis T. The ubiquitin-proteasome myeloma as a disease model. Mayo Clin
Quality-of-life (QOL) and clinical benefit as- pathway is required for processing the Proc 2003;78:1098-109.
sessment in patients with relapsed and re- NF-kB1 precursor protein and the activa- Copyright © 2004 Massachusetts Medical Society.
fractory multiple myeloma (MM) treated tion of NF-kB. Cell 1994;78:773-85.
with bortezomib. Prog Proc Am Soc Clin 110. Hideshima T, Chauhan D, Richardson
Oncol 2003;22:582. abstract. P, et al. NF-kB as a therapeutic target in mul-

n engl j med 351;18 www.nejm.org october 28, 2004 1873

Downloaded from www.nejm.org on March 14, 2005 . This article is being provided free of charge for use in Colombia.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.

También podría gustarte