Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Multiple Myeloma

Seema Singhal* and Jayesh Mehta†


Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Robert H. Lurie
Comprehensive Cancer Center, Northwestern University, Chicago, Illinois

Multiple myeloma is a malignant disease characterized by plasmacytosis, paraprotein production, bone lesions, hypercalce-
mia, susceptibility to infections, and renal impairment. The underlying pathophysiologic phenomena of the clinical features
include suppression of humoral- and cell-mediated immunity, elevation of IL-6, abnormalities of the bone marrow microen-
vironment, and increased osteoclastic activity. Overwhelming predictors of prognosis include albumin, ␤2-microglobulin, and
chromosomal karyotype. With modern, intensive therapy including autologous hematopoietic stem cell transplantation, the
median survival is approximately 5 yr. The disease is incurable and eventually relapses; requiring salvage therapy. The
development of newer agents such as thalidomide, bortezomib, and lenalidomide— drugs that interfere with several of the
complex pathophysiologic steps— has improved the outlook of relapsed disease significantly. Current studies are directed at
exploring the use of these novel agents earlier in the course of therapy, development of newer targeted therapies, and the use
of gene expression profiling to individualize therapy.
Clin J Am Soc Nephrol 1: 1322–1330, 2006. doi: 10.2215/CJN.03060906

M
ultiple myeloma is a hematologic malignancy char- so a decade ago, likely a result of widespread application of
acterized by the clonal proliferation of plasma cells high-dosage chemotherapy with hematopoietic stem cell trans-
in the bone marrow (Figure 1) and, usually, the plantation (HSCT) and new drugs.
presence of a monoclonal Ig in the blood and/or urine. It is the The study of the mechanism of action of these agents has
second most commonly diagnosed hematologic malignancy brought a new insight into the biology of the disease. In addi-
with an annual incidence and prevalence in the United States of tion, as in other areas of medicine, technological advances in
approximately 15,000 and approximately 45,000, respectively. genetic study and bioinformatics have been making significant
The incidence is higher with increasing age (median age at contributions to the understanding of myeloma. This review
diagnosis 67 yr). focuses on the recent progress in the pathophysiology, diagno-
Plasma cell disorders encompass a spectrum including sis, and treatment of myeloma.
monoclonal gammopathy of unknown significance (MGUS),
smoldering or indolent myeloma, and symptomatic myeloma,
in order of increasing tumor burden. The differences between Pathophysiology
these are both quantitative and qualitative; MGUS and smol- There is a growing realization that the evolution of the dis-
dering and indolent myeloma are very slowly proliferative and ease involves sequential and complex changes in both the ma-
relatively stable diseases in contrast to active myeloma. lignant cell and in the surrounding bone marrow microenvi-
Although the disease is largely incurable, the past decade has ronment, including bone. The evolution from MGUS to active
seen dramatic progress in therapy and understanding of its myeloma seems to be a multistep, stochastic progression that
pathophysiology. Since 1998, three new agents with significant can occur over a few months to decades (1).
anti-myeloma activity (thalidomide, bortezomib, and lenalido- The pathogenetic switch or switches that lead to conversion
mide) have been identified. In contrast, the preceding three of normal plasma cell populations to MGUS have not been
decades had been characterized by reliance on two active elucidated. The sequence of events that lead to progression
classes of agents: Alkylating agents (melphalan and cyclophos- from MGUS to active myeloma has been researched intensely
phamide) and corticosteroids (Figure 2). The median survival with a resultant glimpse into the various genetic abnormalities
from diagnosis of patients with symptomatic disease that re- and the bone marrow microenvironment disturbances that exist
quires therapy is approximately 5 yr—an increase from 3 yr or in the various stages of progression. Considerable interpatient
heterogeneity in these disturbances has been shown to exist,
and this has led to proposed new classifications of the disease
Published online ahead of print. Publication date available at www.cjasn.org. on the basis of differences in disease biology.
The tools for the study of the cytogenetic makeup of my-
Address correspondence to: Dr. Seema Singhal, 676 N. St. Clair Street, Suite 850,
Chicago, IL 60611. Phone: 312-695-4129; Fax: 312-695-6189; E-mail: eloma cells mainly have been conventional karyotyping, fluo-
s-singhal@northwestern.edu rescence in situ hybridization, and, more recently, gene expres-

Copyright © 2006 by the American Society of Nephrology ISSN: 1555-9041/106-1322


Clin J Am Soc Nephrol 1: 1322–1330, 2006 Multiple Myeloma 1323

osteoprotegerin result in activation of osteoclasts. Levels of


dickkopf 1 (DKK-1), an inhibitor of Wnt signaling that inhibits
differentiation of osteoblast precursors, also are increased (3,4).
Interactions between the myeloma cell and other cells in the
marrow microenvironment, such as stromal cells and hemato-
poietic stem cells, and also the extracellular matrix activate
multiple signaling pathways, resulting in proliferation/anti-
apoptosis of the myeloma cell. The autocrine/paracrine sur-
vival factors that are involved in these interactions include IL-6,
IGF, VEGF, and TNF-␣ (3). These complex relationships also
contribute to drug resistance, and therapeutic strategies that
simultaneously target both the malignant cell and the microen-
vironment are being designed. It is interesting that only 60% of
patients with myeloma have bone lesions. In these patients, the
resorption of bone seems to be required for fueling disease
progression through release of myeloma survival factors. Other
patients with myeloma do not develop disease-mediated bone
Figure 1. Bone marrow in multiple myeloma showing plasma-
destruction even in advanced stages of the disease. This sug-
cytosis.
gests that the degree of dependence of myeloma cells on the
marrow microenvironment is variable.
sion profiling. There is no characteristic genetic signature that is
diagnostic of the disease. However, activation of one of the
Diagnosis and Investigations
three cyclin D genes has been shown to be present in nearly all Some patients’ myeloma is diagnosed incidentally because of
myeloma cases. Almost half of patients with myeloma have elevated serum protein levels, but most patients present with
translocations that nonrandomly involve the Ig heavy chain symptoms related to anemia, bone lesions, kidney dysfunction,
locus on chromosome 14q32 and one of five well-defined chro- infections, or hypercalcemia. Various blood, urine, bone mar-
mosomal partners: 11q13 (cyclin D1), 6p21 (cyclin D3), 4p16 row, and imaging studies are required to diagnose, stage, and
(fibroblast growth factor receptor 3 and multiple myeloma SET monitor the disease and determine prognosis (Table 1). Table 2
domain), 16q23 (c-maf), and 20q11 (mafB) (2,3). All Ig heavy shows the diagnostic criteria for myeloma (5). A critical element
chain locus translocations except t(11;14) seem to have unfa- of making a correct diagnosis is ensuring that plasmacytosis is
vorable prognosis. Deletions of chromosomes 13 and 17 (17p13; clonal, particularly when other typical features are not present.
the p53 locus) also are unfavorable. Hypodiploid myeloma also
is associated with poorer survival compared with hyperdiploid
myeloma. Classification and Staging Systems
Bone lesions in myeloma are thought to be a result of imbal- The time-honored Durie-Salmon staging system (Table 3)
ance between the two opposing processes: Bone formation by correlates well with tumor burden (6). However, the new in-
osteoblasts and bone resorption by osteoclasts. Myeloma cells ternational staging system (Table 4) (7), based on ␤2-micro-
are thought to increase production of pro-osteoclastogenic cy- globulin and albumin, correlates better with prognosis. The
tokines such as macrophage inflammatory protein 1 (MIP-1␣), translocation and cyclin D classification proposed by Bergsagel
parathyroid hormone–related protein (PTHr-P), vascular endo- and Kuehl (8) has five groups that can be distinguished on the
thelial growth factor (VEGF), and IL-6. An increase in expres- basis of recurrent Ig translocations and cyclin D expression. In
sion of RANKL (receptor activator of NF-␬B ligand) by osteo- a recent proposed molecular classification, seven disease sub-
blasts and a decrease in the level of its decoy receptor types were identified (9). The last two classifications still have

Figure 2. Timeline of treatment evolution in myeloma.


1324 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 1322–1330, 2006

Table 1. Investigations in multiple myelomaa


Investigation Diagnosis Staging Prognosis Monitoring Comments

Hemoglobin ⫹ ⫹ ⫹/⫺ ⫹
Serum calcium ⫹ ⫹ ⫺ ⫹
Serum creatinine ⫹ ⫹ ⫹/⫺ ⫹
C-reactive protein ⫺ ⫺ ⫹ ⫹/⫺
Bone marrow morphology ⫹ ⫺ ⫹ ⫹
Bone marrow cytogenetics ⫺ ⫺ ⫹ ⫹/⫺
Bone marrow FISH ⫺ ⫺ ⫹ ⫹/⫺
Plasma cell clonality ⫹ ⫺ ⫺ ⫹ Flow cytometry or
immunohistochemistry
LDH ⫺ ⫺ ⫹ ⫹/⫺
Skeletal survey ⫹ ⫹ ⫺ ⫹/⫺
Serum free light-chain levels ⫹/⫺ ⫺ ⫺ ⫹/⫺ Essential only if nonsecretory
disease
Serum albumin ⫺ ⫹ ⫹ ⫹/⫺
Serum ␤2-microglobulin ⫺ ⫹ ⫹ ⫺
Serum immunoglobulins ⫹ ⫹ ⫺ ⫹
Serum protein electrophoresis ⫹ ⫹ ⫺ ⫹
Serum immunofixation ⫹ ⫺ ⫺ ⫹
Urine 24-h protein ⫹ ⫹ ⫺ ⫹
Urine 24-h protein electrophoresis ⫹ ⫹ ⫺ ⫹
Urine 24-h immunofixation ⫹ ⫺ ⫺ ⫹
Bone marrow plasma cell labeling index ⫺ ⫺ ⫹ ⫺ Optional
Bone densitometry ⫺ ⫺ ⫺ ⫺ Optional
Skeletal MRI scan ⫹/⫺ ⫹/⫺ ⫺ ⫹/⫺ Optional
PET scan ⫹/⫺ ⫺ ⫺ ⫹/⫺ Optional
a
FISH, fluorescence in situ hybridization; LDH, lactate dehydrogenase; MRI, magnetic resonance imaging; PET, positron
emission tomography; ⫹, essential; ⫹/⫺, utility not unequivocally established; ⫺, not helpful.

to be validated but seem to have promising prognostic and


Table 2. Diagnostic criteria for myeloma from the therapeutic relevance.
Scientific Advisors of the International Myeloma
Foundation (5)
Prognosis
1. ⱖ10% monoclonal plasma cells in the marrow or The international staging system, the translocation and cyclin
biopsy-proven plasmacytoma D classification, and the molecular classification discussed in
2. Monoclonal protein in the serum and/or urine the previous paragraph seem to correlate with survival. How-
3. Myeloma-related dysfunction of at least one organ ever, the international staging system is most easily applicable
system practically because of its simplicity. Other investigations that
Notes are practically feasible and have been shown to correlate with
a. All three criteria must be present. outcome include karyotype and some biochemical parameters.
b. If there is no monoclonal protein (nonsecretory Cytogenetic abnormalities such as partial or complete deletion
disease), then there should be ⱖ30% monoclonal of chromosome 13 and t(4;14) indicate high-risk disease. Other
plasma cells in the marrow or biopsy-proven adverse features include plasmablastic morphology and ele-
plasmacytoma. vated lactate dehydrogenase, plasma cell labeling index, and
c. Typical examples of organ dysfunction are C-reactive protein (10).
hypercalcemia (serum calcium ⬎10.5 g/dl or above
normal), renal insufficiency (serum creatinine ⬎2
mg/dl), anemia (hemoglobin ⬍10 g/dl or ⬎2 g/dl Treatment
Advances in therapy have resulted in improvement in the
less than normal), lytic bone lesions, and
survival of patients with myeloma over the years. Without
osteoporosis.
therapy, the median survival of a patient with active myeloma
d. If a single plasmacytoma or osteoporosis is the
is approximately 6 mo. With oral melphalan-prednisone (MP)
sole organ dysfunction, then the marrow should
therapy, median survival improves to 3 yr. High-dosage ther-
contain ⱖ30% monoclonal plasma cells.
apy with HSCT further improves median survival to 5 yr,
Clin J Am Soc Nephrol 1: 1322–1330, 2006 Multiple Myeloma 1325

Table 3. Durie and Salmon staging system (applicable once the diagnosis of myeloma has been established)
Stage Parameter

Stage I (low tumor burden; all of the following


criteria must be satisfied)
hemoglobin ⬎10 g/dl
serum calcium ⬍12 mg/dl
x-ray No bone destruction (scale 0), or solitary plasmacytoma
low paraprotein production Serum IgG ⬍5 g/dl, or serum IgA ⬍3 g/dl, or urine
light chain ⬍4 g/24 h
Stage II (intermediate tumor burden)
laboratory and radiologic parameters intermediate
between stages I and III
Stage III (high tumor burden; at least one of the
following criteria must be satisfied)
hemoglobin ⬍8.5 g/dl
serum calcium ⬎12 mg/dl
x-ray Advanced lytic bone lesions (scale 3)
high paraprotein production Serum IgG ⬎7 g/dl, or serum IgA ⬎5 g/dl, or urine
serum creatinine ⬍2 mg/dl light chain ⬎12 g/24 h
serum creatinine ⱖ2 mg/dl

Table 4. International staging system general management approach to patients with systemic
plasma cell dyscrasias.
Stage ␤2-Microglobulin (mg/L) Albumin (g/dl)

Ia ⬍3.5 ⱖ3.5 Supportive Therapy


II Fits neither stage I nor stage III Supportive therapy comprises management of hypercalce-
III ⱖ5.5 — mia, skeletal complications, anemia, infections, and pain (13).
a Regular administration of bisphosphonates such as pamidr-
Both criteria must be satisfied.
onate (90 mg once a month) or zoledronate (4 mg once a month)
in patients with skeletal lesions is important (14). These drugs
making this the current standard therapy for myeloma. Tan- arrest and reverse the disturbance of balance between bone
dem autologous transplantation is superior to single in selected formation and resorption. Possible direct and indirect (through
patients. Now the newer agents, such as thalidomide (11,12), effect on the bone marrow microenvironment) anti-myeloma
raise a possible challenge to the established approach of ever- activity of these drugs led to their open-ended use in myeloma.
more aggressive dose escalation. To maximize chances for pro- However, the potential for renal damage and increasing con-
longed survival, it is important to plan therapy for each patient cern about osteonecrosis of the jaw warrants periodic reassess-
in a manner that uses all agents appropriately and sequentially ment of the need for continued bisphosphonates after 2 yr
without adversely affecting future treatment options. The di- rather than the previous approach of long-term therapy. Any
lemma that clinical researchers face is whether to go forward symptoms that involve the jaw should be investigated care-
with trials that combine all available agents to increase chances fully, and a careful dental evaluation and corrective work
of tumor cell kill while increasing the possibility of drug resis- should be performed before starting bisphosphonates or early
tance or to reserve new agents for salvage therapy of relapsed in the course of therapy.
and refractory disease.
The treatment of myeloma comprises disease-specific ther- Disease-Specific Therapy
apy and supportive care. The general principle is to reserve The current standard approach consists of initial induction
disease-specific therapy for active disease. The new diagnostic therapy, consolidation with high-dosage chemotherapy and
criteria for myeloma (Table 2) identify the patients who have autologous HSCT, maintenance therapy, and salvage therapy.
active disease, who almost always need therapy. Definitive Initial Therapy. As HSCT is being used increasingly in
therapy is required when the patient is symptomatic or when myeloma (15,16), the choice of initial therapy is based on
organ dysfunction is present or impending. There is no evi- whether the patient is a transplant candidate. Patients who are
dence that starting definitive therapy in patients with smolder- transplant candidates receive induction therapy that does not
ing or indolent myeloma improves survival. Patients with Du- cause permanent stem cell damage.
rie-Salmon stage I myeloma also can be watched without Patients who are ineligible for HSCT can receive MP or
antitumor therapy for a period of time. Table 5 summarizes the similar therapy. More intensive alkylating agent– based combi-
1326 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 1322–1330, 2006

Table 5. Approach to patients with plasma cell dyscrasias


Supportive Disease-Specific
Diagnosis Monitoring Therapy Therapy

MGUS Annual None None


Smoldering myeloma Every 2 to 3 mo Usually none None
Indolent myeloma Every 1 to 2 mo ⫹/⫺ None
Stage I myelomab Every 1 to 2 mo ⫹/⫺ ⫹/⫺
Stages II to III myelomab At least once a month Yes Yes
a
MGUS, monoclonal gammopathy of unknown significance.
b
Durie-Salmon.

nations improve response rates without prolonging survival. spread adoption of thalidomide-containing induction therapy
However, MP and other alkylating agent– based combinations as the new standard may not be beneficial, because the addition
damage normal hematopoietic stem cells, making stem cell of thalidomide results in higher response rates but increases
collection for HSCT difficult if not impossible and increasing serious toxicity significantly and does not prolong survival. In
the risk for myelodysplastic syndrome. The complete remission a study (20) that compared thalidomide-dexamethasone (TD)
(CR) rate with MP is ⬍5%, and the overall response rate is 40 to with dexamethasone alone, overall response with four cycles
50%. The addition of low-dosage thalidomide to MP in elderly was significantly higher with TD (63 versus 41%; P ⫽ 0.002), but
patients with myeloma improves response rates and event-free CR rates were comparable (4 versus 0%). The incidence of grade
survival (EFS) but increases toxicity significantly compared 3 to 5 deep vein thrombosis (DVT), rash, sinus bradycardia,
with MP (17). Longer follow-up is required to assess effect on neuropathy, and any grade 4 to 5 toxicity within 4 cycles was
overall survival (OS). significantly more common with TD (45 versus 21%; P ⬍ 0.001).
The usual induction therapy for patients who are eligible for OS was identical during the first 2 yr. In another study (21),
HSCT is based on high-dosage dexamethasone or methylpred- patients received intensive induction therapy followed by tan-
nisolone. There is no compelling evidence that regimens that dem autotransplantation and posttransplantation consolidation
combine modestly active intravenous agents with high-dosage and maintenance, with or without thalidomide. Although CR
dexamethasone are better than dexamethasone alone with its rates and 5-yr EFS were better with thalidomide (62 and 56%,
convenience of oral administration (18). Lack of response to respectively) than without (43 and 44%, respectively), the 5-yr
induction therapy does not necessarily signify a poor prognosis OS was similar at 65%. Despite comparable salvage therapy,
because subsequent HSCT can result in substantial cytoreduc- median survival after relapse was only 1.1 yr in the thalidomide
tion (19). group compared with 2.7 yr in the control group (P ⫽ 0.001)
Two recent randomized studies suggested that the wide- because of resistance to all types of salvage therapy in thalid-
omide-treated patients. Severe peripheral neuropathy and DVT
occurred more frequently with thalidomide.
Therefore, a reasonable approach may be to use single-agent
dexamethasone, with consideration being given to the addition
of thalidomide if there is no response to dexamethasone. This
will minimize toxicity and expense without compromising out-
come and likely will reduce the development of resistant dis-
ease. DVT prophylaxis is essential if TD is used, although the
optimum mode of prophylaxis is unclear. The use of other
newer agents, such as bortezomib and lenalidomide, as part of
initial therapy currently is investigational, although the combi-
nation of lenalidomide and dexamethasone seems to be very
active (22).
High-Dosage Chemotherapy. High-dosage melphalan
with autologous HSCT may be administered as consolidation
therapy after induction (early) or as salvage therapy after re-
lapse (late). Two randomized studies showed higher response
rates and prolongation of EFS and OS with early HSCT (15,16),
Figure 3. Overall and event-free survival of 451 patients who
had myeloma and underwent autologous hematopoietic stem whereas one did not (23). Another randomized study showed
cell transplantation after 200 mg/m2 melphalan at the Royal that deferring HSCT to relapse after initial therapy did not
Marsden Hospital between 1982 and 2001 (courtesy of Profes- compromise OS, although EFS was shorter compared with
sor Ray Powles). early transplantation (24). Quality of life, as measured by the
Clin J Am Soc Nephrol 1: 1322–1330, 2006 Multiple Myeloma 1327

Table 6. Drug therapy of myelomaa


Efficacyb Appropriatenessb
Drug/Combination Toxicity Cost Comments
Initial Salvage Initial Salvage
Therapy Therapy Therapy Therapy

Melphalan-prednisone ⫹ ⫹ ⫹⫹ ⫹ ⫹⫹ ⫹⫹ Initial therapy in patients ineligible for HSCT


Melphalan-prednisone-thalidomide ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ ⫹⫹ ⫹⫹ ⫹⫹⫹ Initial therapy in patients ineligible for HSCT
Pulse dexamethasone ⫹⫹ ⫹ ⫹⫹ ⫹⫹ ⫹⫹⫹ ⫹⫹ Initial therapy in patients eligible for HSCT
VAD and equivalentc ⫹⫹⫹ ⫹⫹ ⫹⫹ ⫹⫹ ⫹ ⫹⫹ Initial therapy in patients eligible for HSCT
Thalidomide ⫹⫹ ⫹⫹⫹ ⫹⫹ ⫹⫹ ⫹/⫺ ⫹⫹⫹ Inadequate data to support use as initial therapy
Thalidomide-dexamethasone ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ ⫹⫹ ⫹⫹⫹ Initial therapy in patients eligible for HSCT
Bortezomib ⫹⫹⫹ ⫹⫹⫹ ⫹⫹ ⫹⫹⫹ ⫹/⫺ ⫹⫹⫹ Inadequate data to support use as initial therapy
Bortezomib-dexamethasone ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ ⫹d ⫹⫹⫹ Initial therapy in patients eligible for HSCT
Lenalidomide ⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹ ⫹⫹⫹ ⫹/⫺ ⫹⫹⫹ Inadequate data to support use as initial therapy
Lenalidomide-dexamethasone ⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹⫹ ⫹d ⫹⫹⫹ Initial therapy in patients eligible for HSCT
Combinations of novel agents ⫹⫹⫹ ⫹⫹⫹⫹ ? ⫹⫹⫹ ⫹/⫺ ⫹⫹⫹ Inadequate data to support use as initial therapy
a
HSCT, hematopoietic stem cell transplantation; VAD, vincristine, doxorubicin, and dexamethasone; ⫹/⫺, questionable; ⫹
to ⫹⫹⫹, increasing magnititude; ?, no data.
b
Efficacy and appropriateness ratings for salvage therapy assume no previous exposure to that drug/combination or
exposure with good response and relapse off therapy with that drug/combination.
c
VAD equivalents include VAMP (vincristine, doxorubicin, and methylprednisolone), C-VAMP (cyclophosphamide and
VAMP), and DVD (pegylated doxorubicin, vincristine, and dexamethasone).
d
A low rating despite high response rates reflects ongoing clinical trials and lack of any follow-up data.

TWiSTT score (time without symptoms or treatment toxicity), Allogeneic stem cell transplantation potentially is curative
was better in patients who underwent HSCT early. Another because of immunologically mediated graft-versus-myeloma ef-
recent study that randomly assigned patients to early HSCT or fects (31), but toxicity compromises survival and remains a
continued standard-dosage chemotherapy with an option to concern (32). Minitransplantation, whereby intensity of the con-
undergo salvage HSCT at relapse showed no benefit for early ditioning regimen is reduced substantially, may overcome the
HSCT in terms of response rates, EFS, or OS (25). problem of toxicity in part by making the procedure safer.
Despite these conflicting reports, the balance of opinion cur- Here, there is much greater reliance on graft-versus-tumor re-
rently is that HSCT after high-dosage melphalan is beneficial actions to eliminate the cancer (33).
(26). Figure 3 shows EFS (17% at 10 yr; median 2.4 yr) and OS Although a proportion of patients are alive disease-free for a
(33% at 10 yr; median 6.2 yr) of 451 patients who had myeloma decade or longer after HSCT with essentially normal quality of
and underwent autologous HSCT after 200 mg/m2 melphalan life (“operational cure” [34]), most patients eventually relapse
at the Royal Marsden Hospital (Surrey, UK) between 1982 and and need salvage therapy. Third and occasionally even fourth
2001. cycles of high-dosage chemotherapy have been used in selected
The Arkansas group pioneered the approach of tandem au- patients. Therefore, it is important to collect enough stem cells
tologous HSCT (27). Long-term follow-up of the Total Therapy
1 study from Arkansas shows 10-yr OS and EFS probabilities of
33 and 15%, respectively (28). Further intensification of the
Total Therapy 1 regimen seemed to improve outcome in the
Total Therapy 2 study (29), but improvement stemmed from
intensified chemotherapy rather than added thalidomide (21).
Whether tandem HSCT is superior to single HSCT remains
contentious. The only published, prospective, randomized
study showed a doubling of the likelihood of OS and EFS at 6
yr for patients who underwent tandem HSCT (30). In a sub-
group analysis, this benefit was confined to patients who did
not achieve at least a very good partial response to the first
transplant. Ongoing studies may be able to answer this ques-
tion definitively; early data from these studies favor tandem
HSCT in terms of EFS, but there is no difference in OS yet. The
similarities in long-term outcome between the Royal Marsden
data (Figure 3) and the long-term follow-up for the Total Ther-
apy 1 group may be at least partly because 131 of the Marsden Figure 4. Overall and event-free survival of 169 patients who
patients eventually underwent a second transplant as therapy had end-stage myeloma and were treated with thalidomide
of relapse. (courtesy of Professor Bart Barlogie).
1328 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 1322–1330, 2006

Figure 5. Approach to treatment for a patient who is eligible for stem cell transplantation.

at the outset for the planned number of transplants as well as adverse effects are sedation, fatigue, constipation, peripheral
for future salvage therapy. neuropathy, autonomic disturbances, and thromboembolic
Administration of posttransplantation maintenance therapy phenomena.
may help to delay disease recurrence. The agents used are Figure 4 shows the remarkable long-term outcome of the
corticosteroids, IFN-␣, and thalidomide. Although the use of original cohort of patients who had relapsed myeloma and
maintenance therapy may improve EFS, OS may not be affected received thalidomide as a single agent initially. Long-term fol-
beneficially because recurrent disease may not be sensitive to low-up of the Arkansas Total Therapy 1 study shows longer
the agent used for maintenance. A greater worry is that recur- postrelapse survival for patients whose disease relapsed in the
rent disease may be resistant to other agents as well (21). thalidomide era compared with those whose disease relapsed
Salvage Therapy. The availability of thalidomide (11,12), earlier (28), suggesting that the use of thalidomide as salvage
bortezomib (35,36), and lenalidomide (37,38) has transformed therapy, in contrast to its use as initial therapy, does seem to
the treatment of myeloma. It now is possible to achieve excel- prolong survival.
lent cytoreduction including CR in patients whose disease re- Bortezomib. Bortezomib, an inhibitor of the 26S proteasome
lapses after extensive previous therapy and achieve prolonged (an intracellular organelle that is responsible for protein deg-
survival after relapse. The appropriate approach for a patient radation), also is effective in 25 to 30% of patients with relapsed
with relapsed disease depends on previous therapy, including myeloma (35). For patients with relapsed disease, bortezomib is
transplantation, response to previous therapy, the nature of the more effective than dexamethasone (36). Combining bort-
disease, age, organ function, bone marrow function, the avail- ezomib with other agents that are active in myeloma, such as
ability of an allogeneic donor, and access to clinical trials of corticosteroids, thalidomide, and low-dosage melphalan, in-
investigational agents (39). Table 6 summarizes the activity and creases its efficacy. The main adverse effects are thrombocyto-
the place of various anti-myeloma agents that currently are penia, gastrointestinal disturbances, and peripheral neuropa-
available for salvage therapy. thy. Bortezomib seems to be particularly effective in patients
Thalidomide. Thalidomide as a single agent is effective in with light-chain disease and causes rapid cytoreduction that
approximately one third of patients with relapsed myeloma necessitates precautions against tumor lysis syndrome in pa-
(11,12). The exact mechanism of action is unknown, but the tients with a high burden of rapidly proliferative disease.
drug is thought to act directly on plasma cells, on the marrow Lenalidomide. Lenalidomide is a structural analog of thalid-
microenvironment, and through cytokines that affect the omide but its in vitro biologic actions are more potent than
growth of plasma cells. Some patients respond to as little as 50 thalidomide. It is remarkably effective in patients with relapsed
mg of the drug, but most require 200 mg or so. The addition of disease; including those who have failed prior thalidomide and
other agents such as dexamethasone or multiagent chemother- bortezomib (37,38). The combination of lenalidomide and dexa-
apy improves response rates further and is effective in patients methasone is superior to dexamethasone alone for relapsed
who have not responded to the agents used singly. The main disease (38). While the 25 mg dose has been formally explored
Clin J Am Soc Nephrol 1: 1322–1330, 2006 Multiple Myeloma 1329

(38), the drug is active at doses as low as 5 mg per day (37). The mass with presenting clinical features, response to treat-
main side effects are myelosuppression, thromboembolic phe- ment, and survival. Cancer 36: 842– 854, 1975
nomena (particularly in combination with dexamethasone), 7. Greipp PR, San Miguel J, Durie BGM, Crowley JJ, Barlogie
and skin rash. B, Blade J, Boccadoro M, Child JA, Avet-Loiseau H, Kyle
Combinations. All of these new agents have been combined RA, Lahuerta JJ, Ludwig H, Morgan G, Powles R, Shimizu
K, Shustik C, Sonneveld P, Tosi P, Turesson I, Westin J:
with one or more conventional agents with varying degrees of
International staging system for multiple myeloma. J Clin
success and toxicity. Similarly, combinations of more than one of
Oncol 23: 3412–3420, 2005
the new agents are being studied. Although still investigational, 8. Bergsagel PL, Kuehl WM: Molecular pathogenesis and a
these are reasonable choices in patients who have no other treat- consequent classification of multiple myeloma. J Clin Oncol
ment options. To maximize chances for prolonged survival, it is 23: 6333– 6338, 2005
important to plan therapy for each patient in a manner that 9. Zhan F, Huang Y, Colla S, Stewart JP, Hanamura I, Gupta
utilizes all agents appropriately and sequentially without ad- S, Epstein J, Yaccoby S, Sawyer J, Burington B, Anaissie E,
versely impacting future treatment options (Figure 5). Hollmig K, Pineda-Roman M, Tricot G, van Rhee F, Walker
R, Zangari M, Crowley J, Barlogie B, Shaughnessy JD Jr:
The molecular classification of multiple myeloma. Blood
The Future 108: 2020 –2028, 2006
Therapeutic options for myeloma have expanded dramati-
10. San Miguel JF, Almeida J, Orfao A: Laboratory investiga-
cally with new agents that are active both singly and in com-
tions. In: Myeloma, edited by Mehta J, Singhal S, London,
bination. With several more novel agents in clinical trials, there Martin Dunitz, 2002, pp 243–268
is hope that outcomes will improve dramatically. Future clini- 11. Singhal S, Mehta J, Desikan R, Ayers D, Roberson P, Ed-
cal trials will be designed with rationale based on preclinical dlemon P, Munshi N, Anaissie E, Wilson C, Dhodapkar M,
drug data and patient selection according to pharmacogenomic Zeddis J, Barlogie B: Antitumor activity of thalidomide in
profiles, with sophisticated, individualized therapy. Specific refractory multiple myeloma. N Engl J Med 341: 1565–1571,
experimental agents, approaches, and targets that are being 1999
pursued are too numerous to list entirely. However, a few 12. Barlogie B, Desikan R, Eddlemon P, Spencer T, Zeldis J,
examples include histone deacetylase inhibitors, heat-shock Munshi N, Badros A, Zangari M, Anaissie E, Epstein J,
protein-90 inhibitors, VEGF receptor tyrosine-kinase inhibitors, Shaughnessy J, Ayers D, Spoon D, Tricot G: Extended sur-
vival in advanced and refractory multiple myeloma after
farnesyl transferase inhibitors, and inhibitors of fibroblast
single-agent thalidomide: Identification of prognostic factors
growth factor receptor 3.
in a phase 2 study of 169 patients. Blood 98: 492– 494, 2001
13. Ludwig H, Fritz E: Supportive therapy. In: Myeloma, edited by
Acknowledgments Mehta J, Singhal S, London, Martin Dunitz, 2002, pp 397–412
S.S. and J.M. are members of the speakers’ bureau of and consultant to 14. Berenson JR, Hillner BE, Kyle RA, Anderson K, Lipton A, Yee
Celgene Pharmaceuticals (manufacturer of thalidomide and lenalidomide) GC, Biermann JS: American Society of Clinical Oncology
and Millennium Pharmaceuticals (manufacturer of bortezomib). clinical practice guidelines: The role of bisphosphonates in
multiple myeloma. J Clin Oncol 20: 3719 –3736, 2002
15. Attal M, Harousseau JL, Stoppa AM, Sotto JJ, Fuzibet JG,
References Rossi JF, Casassus P, Maisonneuve H, Facon T, Ifrah N,
1. Seidl S, Kaufmann H, Drach J: New insights into the patho- Payen C, Bataille R: A prospective, randomized trial of
physiology of multiple myeloma. Lancet Oncol 4: 557–564, autologous bone marrow transplantation versus conven-
2003 tional chemotherapy in multiple myeloma. N Engl J Med
2. Stewart AK, Fonseca R: Prognostic and therapeutic signif- 335: 91–97, 1996
icance of myeloma genetics and gene expression profiling. 16. Child JA, Morgan GJ, Davies FE, Owen RG, Bell SE,
J Clin Oncol 23: 6339 – 6344, 2005 Hawkins K, Brown J, Drayson MT, Selby PJ: High dose
3. Mitsiades CS, Mitsiades N, Munshi NC, Anderson KC: therapy improves the outcome of patients with multiple
Focus on multiple myeloma. Cancer Cell 6: 439 – 444, 2004 myeloma: The MRC Myeloma VII randomized trial. N Engl
4. Tian E, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie B, J Med 348: 1875–1883, 2003
Shaughnessy JD Jr: The role of the Wnt-signaling antago- 17. Palumbo A, Bringhen S, Caravita T, Merla E, Capparella V,
nist DKK1 in the development of osteolytic lesions in mul- Callea V, Cangialosi C, Grasso M, Rossini F, Galli M,
tiple myeloma. N Engl J Med 349: 2483–2494, 2003 Catalano L, Zamagni E, Petrucci MT, De Stefano V, Cec-
5. Durie BGM, Kyle RA, Belch A, Bensinger W, Blade J, carelli M, Ambrosini MT, Avonto I, Falco P, Ciccone G,
Boccadoro M, Child JA, Comenzo R, Djulbegovic B, Fantl Liberati AM, Musto P, Boccadoro M: Oral melphalan and
D, Gahrton G, Harousseau JL, Hungria V, Joshua D, Lud- prednisone chemotherapy plus thalidomide compared
wig H, Mehta J, Morales AR, Morgan G, Nouel A, Oken M, with melphalan and prednisone alone in elderly patients
Powles R, Roodman D, San Miguel J, Shimizu K, Singhal S, with multiple myeloma: Randomised controlled trial. Lan-
Sirohi B, Sonneveld P, Tricot G, Van Ness B: Myeloma cet 367: 825– 831, 2006
management guidelines: A consensus report from the Sci- 18. Kumar S, Lacy MQ, Dispenzieri A, Rajkumar SV, Fonseca
entific Advisors of the International Myeloma Foundation. R, Geyer S, Allmer C, Witzig TE, Lust JA, Greipp PR, Kyle
Hematol J 4: 379 –398, 2003 RA, Litzow MR, Gertz MA: Single agent dexamethasone
6. Durie BGM, Salmon SE: A clinical staging system for mul- for pre-stem cell transplant induction therapy for multiple
tiple myeloma. Correlation of measured myeloma cell myeloma. Bone Marrow Transplant 34: 485– 490, 2004
1330 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 1322–1330, 2006

19. Singhal S, Powles R, Sirohi B, Treleaven J, Kulkarni S, Zangari M, Fassas A, Hollmig K, Pineda-Roman M,
Mehta J: Response to induction chemotherapy is not essen- Shaughnessy J, Epstein J, Crowley J: Total therapy 2 with-
tial to obtain survival benefit from high-dose melphalan out thalidomide: Comparison with total therapy 1—Role of
and autotransplantation in myeloma. Bone Marrow Trans- intensified induction and post-transplant consolidation
plant 30: 673– 679, 2002 therapies. Blood 107: 2633–2638, 2006
20. Rajkumar SV, Blood E, Vesole D, Fonseca R, Greipp PR: 30. Attal M, Harousseau JL, Facon T, Guilhot F, Doyen C,
Phase III clinical trial of thalidomide plus dexamethasone Fuzibet JG, Monconduit M, Hulin C, Caillot D, Bouabdal-
compared with dexamethasone alone in newly diagnosed lah R, Voillat L, Sotto JJ, Grosbois B, Bataille R: Single
multiple myeloma: A clinical trial coordinated by the Eastern versus double autologous stem-cell transplantation for
Cooperative Oncology Group. J Clin Oncol 24: 431– 436, 2006 multiple myeloma. N Engl J Med 349: 2495–2502, 2003
21. Barlogie B, Tricot G, Anaissie E, Shaughnessy J, Rasmussen 31. Mehta J, Singhal S: Graft-versus-myeloma. Bone Marrow
E, van Rhee F, Fassas A, Zangari M, Hollmig K, Pineda- Transplant 22: 835– 843, 1998
Roman M, Lee C, Talamo G, Thertulien R, Kiwan E, 32. Mehta J, Tricot G, Jagannath S, Ayers D, Singhal S, Siegel
Krishna S, Fox M, Crowley J: Thalidomide and hematopoi- D, Desikan K, Munshi N, Fassas A, Mattox S, Vesole D,
etic-cell transplantation for multiple myeloma. N Engl Crowley J, Barlogie B: Salvage autologous or allogeneic
J Med 354: 1021–1030, 2006 transplantation for multiple myeloma refractory to or re-
22. Rajkumar SV, Hayman SR, Lacy MQ, Dispenzieri A, Geyer lapsing after a first-line autograft? Bone Marrow Transplant
SM, Kabat B, Zeldenrust SR, Kumar S, Greipp PR, Fonseca 21: 887– 892, 1998
R, Lust JA, Russell SJ, Kyle RA, Witzig TE, Gertz MA: 33. Crawley C, Lalancette M, Szydlo R, Gilleece M, Peggs K,
Combination therapy with lenalidomide plus dexametha- Mackinnon S, Juliusson G, Ahlberg L, Nagler A, Shimoni
sone (Rev/Dex) for newly diagnosed myeloma. Blood 106: A, Sureda A, Boiron JM, Einsele H, Chopra R, Carella A,
4050 – 4053, 2005 Cavenagh J, Gratwohl A, Garban F, Zander A, Bjorkstrand
23. Blade J, Rosinol L, Sureda A, Ribera JM, Diaz-Mediavilla J, B, Niederwieser D, Gahrton G, Apperley JF: Outcomes for
Garcia-Larana J, Mateos MV, Palomera L, Fernandez- reduced-intensity allogeneic transplantation for multiple
Calvo J, Marti JM, Giraldo P, Carbonell F, Callis M, Trujillo myeloma: An analysis of prognostic factors from the
J, Gardella S, Moro MJ, Barez A, Soler A, Font L, Fontanil- Chronic Leukaemia Working Party of the EBMT. Blood 105:
las M, San Miguel J: High-dose therapy intensification 4532– 4539, 2005
compared with continued standard chemotherapy in mul- 34. Powles R, Sirohi B, Kulkarni S, Treleaven J, Rudin C,
tiple myeloma patients responding to the initial chemo- Sankpal S, Goyal S, Horton C, Millar B, Saso R, Singhal S,
therapy: Long-term results from a prospective randomized Mehta J: Collection of peripheral blood stem cells in newly
trial from the Spanish cooperative group PETHEMA. Blood diagnosed myeloma patients without any prior cytoreduc-
106: 3755–3759, 2005 tive therapy: The first step towards an ‘operational cure’?
24. Fermand JP, Ravaud P, Chevret S, Divine M, Leblond V, Bone Marrow Transplant 30: 479 – 484, 2002
Belanger C, Macro M, Pertuiset E, Dreyfus F, Mariette X, 35. Richardson PG, Barlogie B, Berenson J, Singhal S, Jagan-
Boccacio C, Brouet JC: High-dose therapy and autologous nath S, Irwin D, Rajkumar SV, Srkalovic G, Alsina M,
peripheral blood stem cell transplantation in multiple my- Alexanian R, Siegel D, Orlowski RZ, Kuter D, Limentani
eloma: Up-front or rescue treatment? Results of a multi- SA, Lee S, Hideshima T, Esseltine DL, Kauffman M, Ad-
center sequential randomized clinical trial. Blood 92: 3131– ams J, Schenkein DP, Anderson KC: A phase 2 study of
3136, 1998 bortezomib in relapsed, refractory myeloma. N Engl J Med
25. Barlogie B, Kyle RA, Anderson KC, Greipp PR, Lazarus HM, 348: 2609 –2617, 2003
Hurd DD, McCoy J, Moore DF Jr, Dakhil SR, Lanier KS, 36. Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadt-
Chapman RA, Cromer JN, Salmon SE, Durie B, Crowley JC: mauer EA, Facon T, Harousseau JL, Ben-Yehuda D, Lonial
Standard chemotherapy compared with high-dose chemora- S, Goldschmidt H, Reece D, San-Miguel JF, Blade J, Bocca-
diotherapy for multiple myeloma: Final results of phase III doro M, Cavenagh J, Dalton WS, Boral AL, Esseltine DL,
US Intergroup Trial S9321. J Clin Oncol 24: 929 –936, 2006 Porter JB, Schenkein D, Anderson KC: Bortezomib or high-
26. Singhal S: High-dose therapy and autologous transplanta- dose dexamethasone for relapsed multiple myeloma.
tion. In: Myeloma, edited by Mehta J, Singhal S, London, N Engl J Med 352: 2487–2498, 2005
Martin Dunitz, 2002, 327–347 37. Richardson PG, Schlossman RL, Weller E, Hideshima T,
27. Barlogie B, Jagannath S, Desikan KR, Mattox S, Vesole D, Mitsiades C, Davies F, LeBlanc R, Catley LP, Doss D, Kelly
Siegel D, Tricot G, Munshi N, Fassas A, Singhal S, Mehta J, K, McKenney M, Mechlowicz J, Freeman A, Deocampo R,
Anaissie E, Dhodapkar D, Naucke S, Cromer J, Sawyer J, Rich R, Ryoo JJ, Chauhan D, Balinski K, Zeldis J, Anderson
Epstein J, Spoon D, Ayers D, Cheson B, Crowley J: Total KC: Immunomodulatory drug CC-5013 overcomes drug
therapy with tandem transplants for newly diagnosed resistance and is well tolerated in patients with relapsed
multiple myeloma. Blood 93: 55– 65, 1999 multiple myeloma. Blood 100: 3063–3067, 2002
28. Barlogie B, Tricot GJ, van Rhee F, Angtuaco E, Walker R, 38. Dimopoulos MA, Spencer A, Attal M, Prince M, Harousseau
Epstein J, Shaughnessy JD, Jagannath S, Bolejack V, Gurley JL, Dmoszynska A: Study of lenalidomide plus dexametha-
J, Hoering A, Vesole D, Desikan R, Siegel D, Mehta J, sone versus dexamethasone alone in relapsed or refractory
Singhal S, Munshi NC, Dhodapkar M, Jenkins B, Attal M, multiple myeloma: Results of a phase 3 study (MM-010)
Harousseau JL, Crowley J: Long-term outcome results of [Abstract]. ASH Annual Meeting Abstracts 106: 6, 2005
the first tandem autotransplant trial for multiple myeloma. 39. Singhal S, Mehta J: Treatment of relapsed and refractory
Br J Haematol 135: 158 –164, 2006 multiple myeloma. Curr Treat Options Oncol 4: 229 –237,
29. Barlogie B, Tricot G, Rasmussen E, Anaissie E, van Rhee F, 2003

You might also like