MULTIPLE
MYELOMA (MM) / PLASMA CELL MYELOMA / MULTIPLE PLASMACYTOMA OF BONE / KAHLER
DISEASE (Kahler O. Zur Symptomathologie des Multiplen
Myeloms Beobachtung von Albumosurie. Prag. Med. Wochenschr. 14:33-45 (1889))
(indolent)
Table of contents :
Epidemiology (USA)
:
after age 50; peak at age 60; diagnosed annually in approximately 15,000
new patients in the USA, with a prevalence of approximately 50,000 patientsref;
incidence and mortality rates (per 100,000) 1993-1997 :
|
male
|
female
|
|
African American
|
white
|
African American
|
white
|
incidence |
11.4
|
5.1
|
8.6
|
3.3
|
mortality |
7.4
|
3.6
|
5.4
|
2.4
|
year of diagnosis/death |
1976 |
1977 |
1978 |
1979 |
1980 |
1981 |
1982 |
1983 |
1984 |
1985 |
1986 |
1987 |
1988 |
1989 |
1990 |
1991 |
1992 |
1993 |
1994 |
1995 |
1996 |
1997 |
SEER incidence |
4.2 |
4.1 |
3.9 |
3.9 |
4.0 |
4.0 |
4.3 |
4.3 |
4.4 |
4.2 |
4.3 |
4.8 |
4.4 |
4.3 |
4.6 |
4.9 |
4.8 |
4.5 |
4.5 |
4.4 |
4.5 |
4.4 |
US mortality |
2.5 |
2.5 |
2.5 |
2.6 |
2.6 |
2.6 |
2.7 |
2.7 |
2.8 |
2.8 |
2.8 |
2.9 |
2.9 |
2.9 |
3.0 |
3.0 |
3.0 |
3.1 |
3.1 |
3.2 |
3.1 |
3.1 |
SEER mortality |
2.8 |
2.6 |
2.5 |
2.8 |
2.5 |
2.7 |
2.6 |
2.8 |
2.7 |
3.0 |
2.6 |
2.6 |
2.9 |
3.1 |
2.9 |
3.1 |
3.2 |
3.2 |
3.1 |
3.1 |
3.1 |
3.0 |
1-yr |
65.4 |
70.2 |
70.0 |
69.8 |
69.8 |
69.5 |
73.2 |
72.6 |
69.6 |
72.3 |
72.0 |
75.6 |
73.9 |
71.8 |
72.8 |
75.9 |
69.1 |
72.5 |
72.9 |
73.4 |
69.3 |
|
2-yr |
49.3 |
52.0 |
54.8 |
54.1 |
54.8 |
55.6 |
56.2 |
56.1 |
55.6 |
58.9 |
57.1 |
59.4 |
60.1 |
58.3 |
57.1 |
60.6 |
56.3 |
58.2 |
58.4 |
56.3 |
|
|
3-yr |
36.7 |
38.7 |
42.4 |
43.4 |
44.4 |
43.0 |
44.8 |
44.1 |
44.0 |
46.8 |
44.7 |
48.5 |
47.1 |
44.8 |
44.7 |
48.6 |
45.2 |
45.8 |
47.1 |
|
|
|
4-yr |
31.5 |
30.7 |
33.0 |
33.3 |
34.2 |
35.0 |
37.2 |
36.4 |
34.8 |
36.4 |
35.5 |
38.1 |
39.0 |
36.1 |
38.7 |
38.7 |
34.3 |
36.6 |
|
|
|
|
5-yr |
24.6 |
25.0 |
27.3 |
26.1 |
27.0 |
27.8 |
29.7 |
28.7 |
27.6 |
27.7 |
29.8 |
28.8 |
29.8 |
27.0 |
30.7 |
31.2 |
26.4 |
|
|
|
|
|
6-yr |
19.0 |
21.1 |
21.6 |
20.1 |
21.2 |
23.1 |
21.6 |
23.2 |
21.1 |
22.7 |
23.9 |
23.6 |
24.5 |
21.8 |
24.5 |
24.7 |
|
|
|
|
|
|
7-yr |
15.9 |
17.7 |
17.8 |
18.0 |
18.8 |
16.8 |
17.8 |
17.6 |
18.3 |
19.3 |
19.2 |
19.6 |
18.4 |
17.0 |
19.2 |
|
|
|
|
|
|
|
8-yr |
12.4 |
15.4 |
15.1 |
15.4 |
15.3 |
14.4 |
14.6 |
14.2 |
13.8 |
17.0 |
17.8 |
16.5 |
15.5 |
13.6 |
|
|
|
|
|
|
|
|
9-yr |
10.6 |
12.7 |
13.1 |
12.3 |
13.8 |
12.2 |
12.1 |
12.2 |
11.7 |
13.2 |
13.1 |
14.1 |
12.5 |
|
|
|
|
|
|
|
|
|
10-yr |
9.5 |
11.4 |
12.1 |
10.2 |
11.5 |
9.9 |
11.4 |
10.5 |
9.9 |
11.5 |
12.5 |
13.2 |
|
|
|
|
|
|
|
|
|
|
11-yr |
8.7 |
9.3 |
10.9 |
9.1 |
10.4 |
8.8 |
9.7 |
9.9 |
9.7 |
10.2 |
10.8 |
|
|
|
|
|
|
|
|
|
|
|
12-yr |
8.1 |
8.7 |
10.3 |
8.0 |
9.4 |
7.4 |
8.6 |
9.0 |
8.6 |
9.7 |
|
|
|
|
|
|
|
|
|
|
|
|
13-yr |
6.1 |
8.2 |
8.2 |
6.5 |
7.8 |
6.9 |
8.3 |
8.0 |
7.4 |
|
|
|
|
|
|
|
|
|
|
|
|
|
14-yr |
5.2 |
7.9 |
7.9 |
5.8 |
6.0 |
6.6 |
7.2 |
7.3 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
15-yr |
5.0 |
7.6 |
6.3 |
4.5 |
5.8 |
6.2 |
7.2 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
16-yr |
5.0 |
6.6 |
6.3 |
3.9 |
5.3 |
3.4 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
17-yr |
5.0 |
5.7 |
5.5 |
3.9 |
4.7 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
18-yr |
4.4. |
4.6 |
5.0 |
3.7 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
19-yr |
3.6 |
4.5 |
4.5 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
20-yr |
2.3 |
4.4 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
21-yr |
2.3 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Aetiology :
-
benzene

-
chronic antigenic stimulation (CAS)
-
HHV-8
DNA has been identified in phenotypically normalref
bone marrow and peripheral blood adherent stromal cells (dendritic cells)ref1,
ref2
but not in the neoplastic MM plasma cell population. It has been suggested
that the cytokines expressed by HHV-8 gene, such as vIRF, vIL-8R and vIL-6,
may play a role in the pathogenesis of MM. In detection of HHV-8, nested-PCR
is more sensitive than fluorescence quantitative PCRref.
However, the majority of European investigators failed to find evidence
to confirm this observationref1,
ref2,
ref3.
Different explanations have been offered to account for inconsistencies
in seroepidemiologic and virologic studies searching for an association
of KSHV with MM, including the presence of viral variants. Indeed, Ma et
alref
reported unique KSHV open reading frame (ORF) 65 sequences that were detected
exclusively in patients with MM but not in patients without MM who were
seropositive for KSHV. Interestingly, in some MM patients, a mutation at
the C-terminal end of ORF65 resulted in an extension of the ORF by 33 amino
acids. These sequences were found only in samples from patients with MM
and were suggested as a possible unique target of KSHV serologic immune
responses in such patients, who lack seroreactivity against the "conventional"
ORF65 protein : however these possibilities are unlikely, because very
sensitive RT-PCR analyses, with a detection limit of 1 viral copy in 10
× 103 cells, was performed by using primers specific for
3 different KSHV genesref1,
ref2.
To avoid detection by this RT-PCR, the virus in patients with MM would
have to be changed in all these locations and represent a very distant
relative of KSHV. Moreover, if all those KSHV proteins that have been shown
to be targeted by humoral or cellular immune responses were either altered
or completely absent in the virus associated with MM, the elusive infectious
agent would likely be unrelated to the KSHV described by Chang et al in
1994ref.
Nevertheless, when the recently described ORF65 sequence unique to patients
with MM was included in the ELISPOT analyses no specific responses were
detected in any patient with MM. Although we cannot rule out the possibility
that ongoing viral replication was insufficient to maintain detectable
memory CTL responses against the lytic ORF65 protein, one would expect
to detect at least some responses against the latent gene product of ORF73,
which is required for viral genome persistenceref1,
ref2.
The level of viral replication may be very low and not detectable with
the sensitive RT-PCR method used. Such low-level viral replication could
also lead to an antigenic load insufficient for induction of humoral and
cellular immune responses. It remains unclear whether HHV-8 is consequentially
associated with MM or only a regional opportunistic infection. Controversy
still focuses on the role of specific HHV-8 strains involved in pathogenesis
of MMref
-
adiposity
: RR= 1.5 for BMI, 1.9 for weight, 2.0 for waist circumference, and 1.8
for hip circumferenceref
-
familial MM : RR= 3.23 in first-degree
relatives of female sex and also a 2-fold risk for other hematologic malignancies.
Although evidence for an increased risk for MGUS was not documented in
this series as it has been in other reports, this may be due to the fact
that the relatives of MM patients were not systematically screened for
the presence of paraproteins. Therefore the inference in this report about
the familial risk of MGUS is rather weak. On the other hand, the authors
identified 8 families in which the propositus had myelomatosis and at least
one relative had MGUS and another gammopathyref1,
ref2.
Pathogenesis :
all patients with MM are felt to evolve from a MGUS/SMM stage, although
in many MM patients these premalignant stages are unrecognized clinically
due to their asymptomatic nature. A prior history of MGUS or SMM has no
impact on the prognosis of MM. MM plasma cells, which originate from postfollicular
B cells, are characterised by complex chromosomal aberrations. Among the
earliest genetic events are
-
translocations of the IgH gene locus, which leads to dysregulation
of oncogenes at translocation partner regions (cyclin
D1
at 11q13, FGFR3
/MMSET
at 4p16.3, c-MAF
at 16q23, and cyclin D3
at 6p21)
-
deletions of 13q14 (assessed with double color FISH), the site of
a putative tumour suppressor gene, which is an adverse prognostic indicator
(worse EFS and OS, higher relapse rate patients with deletion 13q
had a nearly 2 times higher risk of non surviving (HR: 1.94; 95% CI: 0.95-3.98,
p=0.07) after dose-reduced allogeneic stem cell transplantation)
-
epigenetic changes and activating mutations of oncogenes are usually
associated with disease progression :
-
mutations of N-RAS
and K-RAS
study |
KRAS2 mutations (expecially codon 12) |
NRAS mutations (expecially mutations at codon 61
in a subset representing 12-100%ref) |
medullary disease, at diagnosis [at relapse] (%) |
extramedullary disease |
medullary disease, at diagnosis [at relapse] (%) |
extramedullary disease |
Neri et alref
(1989) |
2/43 [2/13] (7) |
NA |
11/43 [4/13] (26) |
NA |
Paquette et alref
(1990) |
0/17 (0) |
NA |
2/17 (11) |
NA |
Matozaki et alref
(1991) |
0/15 (0) |
NA |
4/15 (26) |
NA |
Tanaka et alref
(1992) |
0/10 (0) |
NA |
5/10 (50) |
NA |
Portier et alref
(1992) |
2/13 (15) |
6/15 (40) |
2/13 (15) |
6/15 (40)* |
Corradini et alref
(1993) |
0/77 (0) |
1/13 (7) |
7/77 (9) |
3/13 (29)* |
Millar et alref
(1995) |
0/18 (0) |
NA |
0/18 (0) |
NA |
Liu et alref
(1996) |
23/139 (16.5) |
NA |
29/129 (23) |
NA |
Bezieau et alref
(2001) |
35%
|
|
25%
|
|
-
changes in c-MYC

-
SHP1 methylation leading to epigenetic activation of the Jak/STAT pathwayref
-
c-MAF
is overexpressed in 50% of MM : the expresson of integrin b7
(which binds E-cadherin
on the surface of stromal cells upregulating expresion of VEGF, which is
important for MM cell growth and enhances the proliferation through the
production of cyclin D1
)
and the cell-cycle regulator cyclin
D2
,
and the chemokine receptor CCR1
is increased in the presence of c-MAFref



Bone-marrow stromal cells support growth and survival of MM cells via
various cytokines. Osteoclast activity factors (OAF = IL-1b
,
TNF-a
,
TNF-b
,
... in particular CCL-3 / MIP-1a
)
and imbalances between RANKL and osteoprotegerin are major factors for
the development of MM bone disease. MM cells secrete dikkopf
homolog 1 (DKK1) which inhibits the differentiation of osteoblastic
precursors in vitroref.
Frequent expressions of TLRs were detected in cell lines from MM patients
(minimum 6 TLRs in each). In comparison, only few TLRs (mainly TLR1
and or RP105
)
were found expressed in PCs from BM of healthy donors. In addition, TLR-specific
ligands induce increased proliferation and survival of the MM cell lines,
partially due to an autocrine IL-6 production. Importantly, also PC from
MM patients proliferates in response to TLR-specific ligandsref.
In MM patients, a large fraction of peripheral blood CD8+
cells display the phenotype of chronically activated memory T cells(CD25-28-38+45RO+
HLA-DR+).
Unlike normal CD8+ T cells, in which CD94 is assembled with
glycoproteins of the NKG2 family to form functional receptors with activating
or inhibitory properties, most CD8+94+ MM T cells
are devoid of both the NKG2-A and NKG2-C glycoproteins detected in the
inhibitory or activating form respectively and do not express a functional
CD94 receptor. Thus, their ability to 'fine-tune' an appropriate immune
response against tumour cells can be impairedref.
The degree of TCR diversity is similar in age-matched normal donors and
MGUS, but progressively decreases from MGUS to MM at diagnosis and then
to MM in remission. After high-dose chemotherapy and autologous peripheral
blood progenitor cell infusion, on average, 33% of the total repertoire
in both the peripheral blood (PB) and bone marrow (BM) consist of naive
or memory CD8+ T cells expressing oligoclonal TCRb
transcripts. There is a significant decrease of PCK-a
in MM T cells; however, neither this decrease nor the heterogenous levels
of the other T-cell signalling molecules are clearly correlated with prognosis,
duration of tumour exposure, and disease statusref.
Fas+ T cells are significantly higher, whereas bcl-2+
T cells are significantly lower in MM patients than in the controls, leading
to enhanced susceptibility to apoptosisref.
In bone marrow the percentage of CD4+ is profoundly reducedref.
Severe and long-lasting disruption of TcR diversity in human myeloma may
affect the clinical outcome of vaccine-based strategies delivered at the
stage of MRDref
: the immune competence status of MM patients is still susceptible to specific
immunization after high-dose chemotherapy and PBPC transplantation and
generates Id-specific T-cell proliferative responsesref
whose ability to reduce the relapse rate of patients with MRD has not been
determined yet.
Several studies have shown that bone marrow-derived endothelial
cells (ECs) may contribute to tumor angiogenesisref1,
ref2,
ref3,
and that in the peripheral blood of cancer patients there is an increased
amount of circulating
ECs (CECs)
ref
(very low number of CECs in peripheral bloodref1,
ref2)
that may participate to vessel formationref.
Microvascular
ECs in B-cell lymphomas are in part tumor-related, reflecting a novel aspect
of tumor angiogenesisref.
All together, these observations suggest that tumors can elicit the sprouting
of new vessels from existing capillaries through the secretion of angiogenic
factorsref
and that, in some cases, cancer cells can also mimic the activities of
ECs by participating in the formation of vascular-like networksref1,
ref2.
In MM, the proliferation and survival of neoplastic plasma cells is regulated
by microenvironmental bone marrow factors and, to this extent, neoangiogenesis
is thought to have a key role in the pathogenesis and progression of the
diseaseref.
It has been shown that in patients with MM, ECs differ markedly from umbilical
vein ECs, their quiescent counterpart, with regard to the secretion of
growth factors, growth properties, genetic profile, and structural featuresref.
In
patients with MM, the level of CECs, which comprise mature ECs and
endothelial
progenitor cells (EPC)
,
were higher than in controls and correlated positively with serum M protein
and b2-microglobulin, thereby representing
a vascular marker that reflects tumor mass and prognosisref.
In addition, a correlation was documented between the level of CECs/EPCs
and response to thalidomide
treatmentref,
suggesting an antiangiogenetic mechanism of thalidomide action. In 5 MM
patients with 13q14 deletion, CECs carried the same chromosome aberration
as the neoplastic plasma cells (11%-32% of CECs with 13q14 deletion;
CD38-138-144+146+
UEA-1 lectin+,
VWF+, VEGFR-2+) and presented the same immunoglobulin
gene rearrangement as MM plasma cells. Most of the CECs displayed immunophenotypic
features of endothelial progenitor cells (CD133+, a marker gradually
lost during endothelial differentiation and absent on mature endothelial
cells). To the contrary, in 3 patients with MGUS and 13q14 deletion, CECs
were cytogenetically normal and had a mature immunophenotype. These findings
suggest a possible origin of CECs from a common hemangioblast precursor
that can give rise to both plasma cells and endothelial cells, as suggested
by evidence coming from studies in patients with chronic
myeloid leukemia
ref,
and point to a direct contribution of MM-derived CECs to tumor vasculogenesis
and possibly to the spreading and progression of the diseaseref.
It has been shown that VEGFR-2 is the only marker shared by CECs and plasma
cells, and that MM plasma cells are negative at mRNA and protein levels
for most of endothelial cell markers including factor VIII-related antigen,
VE-cadherin, and UEA-1ref.
Cell fusion, in our study, seems unlikely because the fusion of MM plasma
cells and ECs should result in a tetraploid karyotype and, in our patients
with MM, all CECs contained a normal diploid copy number of chromosome
10. It seems also unlikely that, as observed in solid tumors, our findings
could reflect an inherent cytogenetic instability of tumor endothelial
cells, because FISH results were not consistent with the heterogeneous
cytogenetic profile of ECs observed in solid tumorsref.
Disease progression :
variable
|
initial phase
|
medullary relapse
|
extramedullary relapse
|
site of myeloma cell accumulation or proliferation |
bone marrow |
bone marrow |
blood, pleural effusion, skin, many other sites |
growth fraction (rate of atypical cells proliferating in the bone marrow) |
< 1% |
>= 1% (1-95%) |
>= 1% (1-95%) |
genetic or oncogenic events |
deregulation of c-myc. Illegitimate switch recombination |
N-ras and K-ras mutations |
p53 point mutations |
phenotypic changes |
CD19 loss, CD56 overexpression |
CD28 expression, LFA-1 and VLA-5 loss |
CD28 expression, CD56 loss |
cytologic changes |
detectable plasmablastic compartment in 15% of cases |
plasmablastic compartment growing |
major plasmablastic compartment |
circulating malignant plasma cells |
< 1% |
increasing |
increasing |
Functional FoxP3+ Treg
cells
of naive, central, and effector memory phenotype as determined by
CCR7 and CD45RA expression are significantly expanded. Low frequencies
of TRECs in naive Treg cells in both healthy controls
and MM patients point to peripheral expansion as the prominent mechanism
of increased frequencies of naive Treg cells in these
cancer patients. These findings strongly suggest that the increase of functional
Treg cells in cancer patients is a response to the process of
malignant transformationref.
Cytology : in contrast to normal plasma
cells, myeloma cells are often immature and may have the appearance
of plasmablastsref.
They usually are CD19- CD56brightref,
CD38+, and syndecan-1, and they produce very low amounts
of immunoglobulins (a few picograms per cell per day)ref.
In almost all patients, the myeloma cells are aneuploid (more often hyperdiploid
than hypodiploid)ref,
and their chromosomes have many numerical and structural abnormalities,
mainly on chromosomes 13 (13q-) and 14 (14q+)ref.
These genetic abnormalities may prevent the differentiation and normal
death of the myeloma cells, which continue to proliferate and accumulate
in the bone marrow. The morphologic immaturity (cells taking the form of
plasmablasts)ref1,
ref2,
hypodiploidyref,
and the 13q- and 14q+ abnormalitiesref
correlate with the resistance to treatment and short survival characteristic
of aggressive disease. The somatic mutations of the immunoglobulin genes
of myeloma cellsref
indicate that the putative myeloma-cell precursors are stimulated by antigens
and are either memory B cells or migrating plasmablasts. The stability
of the mutationsref
and of the antigenic properties (the idiotype) of the myeloma protein during
the course of the disease has clinical implications. The mutations, which
are molecular signatures of the neoplastic clone, might be useful for detecting
residual myeloma cells after chemotherapy. Vaccination with the myeloma
idiotype of a monoclonal immunoglobulin is a potential means of immunotherapy.
Myeloma cells proliferate slowly in the marrow. < 1% of them divide
at any one timeref,
and they do not differentiate completely. The cause of this failure to
differentiate is unknown, but translocations between 14q32 and its chromosome
partners (chromosomes 11, 6, 16, 9, 18, and 8)ref1,
ref2
and deregulation of the c-myc oncogeneref
may be important. The growth fraction of the tumor is high (> 20%) in relapses
in bone marrow, but especially so in relapses outside the bone marrowref.
Point mutations of the N-ras and K-ras oncogenes have been found in relapses
in marrowref,
and point mutations of p53 have been identified in extramedullary relapses
of myelomaref.
Cytologicref1,
ref2
and phenotypicref
changes have also been associated with both these types of relapses. Although
p53 mutations are rarely seen at the time of diagnosis, N-ras and K-ras
mutations are apparent in up to 15% of patients given a new diagnosis of
MM. K-ras mutations are associated with shorter survivalref.
MM is usually thought to be confined to the bone marrow, but recent studies
have noted circulating myeloma cells in many patientsref.
The absolute number of these cells correlates with disease activity and
predicts the progression of disease in SMM. Circulating myeloma cells may
disseminate the tumor within the bone marrow and elsewhere. IL-6 is essential
for the survival and growth of myeloma cellsref1,
ref2,
which express specific receptors for this cytokine. The IL-6R
has 2 polypeptide components: the a chain (composed
of the glycoprotein subunit gp80, or IL-6Ra)
and the chain, a transducer element (gp130). IL-6 belongs to a family
containing 5 other cytokines that use gp130 as a transducer: oncostatin
M
,
leukemia
inhibitory factor
,
IL-11
,
CNTF
,
and cardiotrophin 1
.
IL-6 was initially found to be a growth factor for myeloma cellsref1,
ref2,
but recently it was also shown to promote the survival of myeloma cells
by preventing spontaneous or dexamethasone
-induced
apoptosisref.
These data from in vitro studies suggest that IL-6 promotes both
tumor growth and resistance to dexamethasone in vivo. The beneficial
effects of therapy with murine anti–IL-6 monoclonal antibodies in some
patients with advanced MM also support this suppositionref.
There is increasing evidence that IL-6 is not an autocrine growth factorref,
but
the product of other cells in the microenvironment of the marrowref.
Indeed, myeloma cells can stimulate stromal cells and bone cells to release
large amounts of IL-6ref.
Various membrane proteins on myeloma cells (and on their normal neighbors
in the marrow), in addition to soluble factors produced by normal cells
in the microenvironment, help induce the production of IL-6ref.
The increased levels of IL-6 in the serum of patients with MM can
be explained by the overproduction of IL-6 in the marrow. Myeloma cells
shed the soluble form of IL-6R, which can amplify the response of myeloma
cells to IL-6ref.
IL-6Ra
is present in high amounts in the serum of patients with myeloma, especially
those with a poor prognosis. The IL-6 system also has a role in the
pathogenesis of bone lesions in MMref.
IL-6, sIL-6Ra, and IL-1 activate osteoclasts
in the vicinity of myeloma cells and thus provoke bone resorption.
-
micromolecular myeloma / light
chain myeloma / Bence Jones myeloma (10%) : only monoclonal Ig light
(L) chains are produced. Such chains are found minimally in plasma
as they can pass glomerular filters, precipitate within renal tubules and
be recovered in urine as dimers (Bence Jones (BJ) protein (BJP)
(Bence Jones Henry. On a new substance occurring in the urine of a patient
with mollities ossium. Phil.Transact.Roy.Soc 138:55-62 (1847))), leading
to
glomerulonephritis
and renal failure
.
It has unusual solubility properties : on heating it precipitates at 50°-60°C
and redissolves at 90°-100°C, and on cooling it again precipitates
and redissolves (Bence Jones reaction)
-
non-secretory myeloma (0.1-5%)
: absence of detectable M-protein in serum and urine. The presenting
features of nonsecretory myeloma are similar to those in patients with
a detectable M-protein, except for the absence of renal function impairment.
The response to therapy and survival of patients with nonsecretory myeloma
are similar to those of patients with measurable M-protein. Coexistence
of nonsecreting and nonproducing light chain MM has been reportedref
-
non-producing myeloma : no cytoplasmic
immunoglobulin chains are synthetized
Localizations
:
-
plasmacytoma (< 5% of patients with
a plasma cell dyscrasia) present with a single bone (SBP) or extramedullary
plasmacytoma (EMP) without evidence of systemic disease (normocalcemia,
absence of anemia, preservation of uninvolved immunoglobulins, or renal
disease attributable to myeloma). Diagnosis requires biopsy confirmation
of a monoclonal plasma cell infiltrate from a single site. The treatment
of choice for both entities is radiotherapy given with curative intent
(> 4000 cGy) resulting in long term disease-free survival in approximately
30% of patients with SBP and 65% of patients with EMP.
-
solitary plasmacytoma
of bone (SPB) / bone localized or solitary myeloma : diagnosis requires
-
biopsy-proven monoclonal plasma cell infiltration of a single lytic bone
lesion
-
no additional lesions on bone survey : the role of MRI
of the thoracic and lumbosacral spine in the diagnosis of SBP was prospectively
evaluated by Moulopoulos et al in 12 consecutive patients with SBP : MRI
detected additional abnormalities in 4 patients who, after radiotherapy
for the solitary lesion (of curative intent), had persistent paraprotein
(> 50% of the pretreatment value), compared with only 1 of 6 similarly
treated patients without an abnormality by pretreatment spinal MRIref.
In a follow-up report of 23 patients with thoraco-lumbar spine disease
from one center, 7 of 8 patients who had SBP by radiographs alone developed
MM, compared with only 1 of 7 patients with only 1 lesion confirmed by
MRIref.
Thus, most investigators agree that a negative MRI of the thoraco-lumbar
spine is a prerequisite for the diagnosis of SBP
-
absence of clonal plasma cells on a random marrow sample : 33% of patients
considered to have a SPB according to standard criteria have bone marrow
abnormalities consistent with MM on MRI
ref
-
no evidence of systemic MM (normocalcemia, absence of anemia or renal disease
attributable to MM)
-
presence of monoclonal protein (M protein) in the serum or urine has been
noted in 24-72% of patients in various seriesref1,
ref2,
ref3.
Among 63 consecutive previously untreated patients with SBP, 62% had a
serum M protein, 13% had only Bence Jones protein (BJP), and 25% had non-secretory
diseaseref.
Paraprotein values were usually very low, with only 11 of 37 patients with
a serum M protein > l g/dL (high value 2.2 g/dL) and the highest urine
BJP was 0.7 g/day
-
serum immunoglobulin free light chain (FLC) assays have provided a measurable
parameter to follow in approximately 65% of patients previously diagnosed
with "non-secretory" MM by standard electrophoretic studiesref.
It is likely that these assays will also be useful in a similar percentage
of patients with SBP. Like MRI, this would improve the precision of monitoring
SBP and provide more sensitive identification of patients who achieve complete
disappearance of paraprotein after radiation therapy and are most likely
to be cured. An abnormal FLC ratio at diagnosis may identify risk of progression
to MM in patients with solitary bone plasmacytoma (SBP). In the cohort
of 116 patients, 43 have progressed to MM; with a median time to progression
of 1.8 years. The FLC ratio was determined in all 116 patients on serum
collected at time of diagnosis, and was abnormal in 54 patients (47%).
An abnormal FLC ratio was associated with a higher risk of progression
to myeloma (p=0.039). The risk of progression at 5 years was 44% in patients
with an abnormal serum FLC ratio at diagnosis compared to 26% in those
with a normal FLC ratio. 1-2 years following diagnosis, a persistent serum
M protein 0.5 g/dL was an additional risk factor for progression. Risk
stratificationref
:
-
patients with a normal FLC ratio at baseline and M protein <0.5 g/dL
1-2 years following diagnosis (low-risk; n=31) => 5-yr PFS = 13%
-
either risk factor abnormal (intermediate-risk, n=26) => PFS = 26%
-
both an abnormal FLC ratio and M protein 0.5 g/dL (high-risk, n=18) =>
PFS = 62%
-
levels of uninvolved immunoglobulins are usually preserved, and in our
series of 63 patients, only 3 had suppressed levels. These 3 patients had
disease progression at 12, 18 and 74 months, indicating the presence of
systemic diseaseref.
Therapy : radiotherapy
with curative intent is the treatment of choice, resulting in local control
in > 80% of patients and prolonged cause-specific disease-free survival
in approximately 35% of patients with SBP. Strict dosing guidelines are
difficult to recommend due to the limited number of patients and the absence
of prospective phase I-II and randomized studies. Despite this, some recommendations
are reasonable based on multiple single institution studies. Tsang et al
report no dose-response above 35 Gy in 32 patients; however, a retrospective
review by Mendenhall et al noted that among 81 patients treated with >
40 Gy of radiation, there was only 6% local progression of disease, which
was superior to 31% for those who received < 40 Gyref1,
ref2.
Similarly, no local failure was seen for doses > 50 Gyref.
While no dose-response was noted by Tsang et al, size of the tumor appeared
predictive for local recurrence. That report indicated that SBP < 5
cm was 100% controlled for local recurrence, while nearly 40% of patients
with bulky disease (>= 5 cm) had local progression or recurrence of diseaseref.
Outcome after treatment for solitary plasmacytoma (selected studies) :
series
|
no. of patients |
radiotherapy dose (Gy) |
10-year DFS (%) |
OS (yrs) |
10-year OS (%) |
Wilderref |
60 |
3-70 |
38 |
11 |
- |
Frassicaref |
46 |
< 12-70 |
25 |
9.3 |
- |
Tsangref |
32 |
< 30-0 |
36 |
10 |
- |
Bolekref |
27 |
28.3-60 |
46 |
10 |
- |
Thus, in the absence of a large cooperative group study, our practice has
been to recommend 45 Gy in 25 fractions over 5 weeks. Recently, evidence-based
guidelines of the United Kingdom Myeloma Forum (UKMF) recommend radiotherapy
of at least 40 Gy in 20 fractions and ports that encompass the tumor volume
plus a margin of at least 2 cm beyond disease detectable by MRIref.
For bulkier disease (> 5 cm), a higher dose (up to 50 Gy in 25 fractions)
was recommended.Surgical resection of SBP is rarely necessary, but occasional
patients may require decompressive laminectomy in the presence of cord
compression. When possible, an anterior approach is preferred and radiotherapy
is generally delayed, but ports may be somewhat compromised due to hardwareref.
Adjuvant chemotherapy has been administered with inconclusive results.
Although some studies have found that adjuvant therapy may prevent or delay
progression to myeloma, most have noted no benefit with the early administration
of chemotherapyref1,
ref2,
ref3.
More recently, even myeloablative therapy with stem cell support has been
evaluated in high-risk patients with solitary bone plasmacytoma, but results
are too premature to draw any conclusions given the long natural history
of this diseaseref.
It is unclear whether the risk of early therapy for SBP may predispose
to development of either resistant plasma cell clones or to secondary leukemia.
Thus, given the lack of consistent data proving benefit from chemotherapy,
we and others believe that there is no current role for adjuvant chemotherapy
in the initial treatment of SBP.
Prognosis : while the majority of patients
with solitary plasmacytoma of bone develop myeloma after a median of 2-3
years, the overall median survival of 7-12 years is longer than for patients
in early phases of symptomatic myeloma. Approximately 15-45% of patients
remain disease free at 10 years, and although the majority of these appear
to be cured, rare late recurrences have been reported. In our experience,
prolonged stability ensued only in patients with complete disappearance
of paraprotein by one year, indicating the absence of occult disease outside
the radiotherapy port, in contrast to patients with either persistence
of paraprotein (after 1 year) or non-secretory disease in whom myeloma
usually evolvedref.
Others have reported that neurologic problems associated with SBP, spinal
disease, soft tissue masses, bulky disease (>= 5 cm), age > 55 years, radio-therapy
dose, and M protein level were important prognostic factorsref1,
ref2,
ref3,
ref4,
ref5,
ref6,
ref7,
ref8,
ref9,
ref10,
ref11.
In a series of 63 patients researchers were unable to confirm any of these
factors, although no patient with a serum M protein > l.0 g/dL had complete
disappearance of paraprotein.
Myeloma-free survival (A) and cause-specific survival (B), both in
terms of response of myeloma. (M) protein in the blood and/or urine to
radiotherapy, for solitary bone plasmacytoma patients.
As further advances in diagnostic testing are evaluated (PET scans,
free light chains) occult disease should be more easily recognizable. Consequently,
SBP will be diagnosed less, complete disappearance of paraprotein should
occur more frequently and the fraction of cured patients should rise.
-
solitary extramedullary
or extraosseous plasmacytoma (EMP) / soft tissue myeloma is less common
than SBP and occurs when there is soft tissue infiltration of clonal plasma
cells. There should be no evidence of bone destruction or occult disease
elsewhere. EMP must be distinguished from reactive plasmacytoma, plasma
cell granuloma and lymphoma (MALT, marginal zone, and immunoblastic)ref1,
ref2.
This is probably best accomplished by phenotypic studies positive for CD38
and monoclonal cytoplasmic light chain expression of malignant plasma cells
obtained by biopsy or fine needle aspiration of the solitary lesion. Similar
to SBP, there should be no evidence of clonal marrow plasmacytosis. Approximately
85% of lesions occur in the head and neck mucosa, and underlying bone involvement,
particularly in the sinuses, may be noted. Gastrointestinal involvement,
although significantly less common, is the next most frequent site and
other areas of involvement reported infrequently include lung, bladder,
thyroid, testis, ovary, and tonsil among othersref1,
ref2,
ref3.
There are limited data regarding the diagnosis and staging of this disorder,
and while CT or MRI is required to document the extent of the solitary
lesion, no prospective data exist regarding the role of spinal MRI in staging
this entity, but similar to SBP, we find it useful for the accurate staging
of EMP. < 25% of patients have evidence of a low level of monoclonal
protein in serum or urine by electrophoresis and/or immunofixation, and
(similar to our experience with SBP) we require normal levels of uninvolved
immunoglobulins to confirm the absence of occult disease elsewhereref.
Although current experience is minimal, free-light chain assays should
also prove useful in monitoring such patients, particularly those classified
with non-secretory features. In addition, patients should have no sign
of underlying myeloma by bone survey and chemistries should reveal no abnormalities
attributable to plasma cell dyscrasia.
Therapy : like SBP, EMP are highly sensitive
to radiotherapy
with nearly all patients successfully achieving local control (80%-100%)
and approximately the 50-65% of patients remaining free of disease longer
than 10 yearsref1,
ref2,
ref3,
ref4,
ref5.
Due to small patient numbers and historical retrospective analyses over
many decades, no firmly established criteria for treatment exist. Similar
to SBP, Tsang et al reported successful local control for nearly all patients
(13 of 14) treated with 35 Gy with only 1 failure in a patient with disease
> 5 cm; others also confirmed less satisfactory control in sites with bulky
disease.8 Median doses of radiation in most studies ranged from 35 to 45
Gy and in our own experience, there appears to be little value for doses
beyond 50 Gy (myeloma developed in 1 of 5 patients who received 40-49 Gy
versus 4 of 15 patients treated with 50-60 Gy)ref.
The utility of prophylactic irradiation of local lymph nodes is unclear
as there have been excellent results with elective inclusion (but with
increased morbidity), as well as similar results with inclusion of draining
lymph nodes only when clinically involved (or for Waldeyer's ring involvement,
which is considered at high risk for local recurrence)ref1,
ref2,
ref3,
ref4.
series |
no. of patients |
radiotherapy dose (Gy) |
10-year DFS (%) |
OS (yrs) |
10-year OS (%) |
Galieniref |
46 |
40-75 |
78 |
- |
80 |
Liebrossref |
22 |
40-60 |
56 |
- |
50 |
Brinchref |
18 |
28-60 |
80 |
- |
76 |
Chaoref |
16 |
40-50.4 |
75 |
- |
54 |
Tsangref |
14 |
< 30-50 |
84 |
- |
65 |
Hollandref |
14 |
16.11-62 |
64 |
- |
NA |
A group reported 100% local regional control in 5 of 7 patients with tumors
in the oral cavity, oral/nasopharynx, larynx, or parotid treated with elective
nodal radiotherapyref.
Because of these excellent results, we currently recommend elective radiation
of nodal areas, but not for maxillary sinus or nasal cavity involvement.
Current evidence-based recommendations by the United Kingdom Myeloma forum
are otherwise similar to our own recommendations and those for SBPref.
Their recommendations include a radiotherapy dose of 40 Gy in 20 fractions
for tumors < 5 cm and up to 50 Gy in 25 fractions for tumors >= 5 cm
with at least a 2 cm margin encompassing the primary tumor. If cervical
nodes are involved (or in Waldeyer's ring tumors), these should be included
in the radiotherapy field. MM-free survival for patients with extramedullary
plasmacytoma (EMP) (A) and overall and cause specific survival for all
patients (B).
Because EMP is a highly radiosensitive tumor, surgical procedures of
the head and neck are not recommended, but surgery may be considered for
other sites of disease, such as the gastrointestinal tract. This approach
is supported by a literature review by Alexiou et al, in which patients
with sites other than the head and neck received either surgery, radiation,
or a combined-modality treatmentref.
There was no difference between the 3 arms, suggesting that either surgery
or radiotherapy is reasonable for such patients. Considering the high cure
rate of EMP with radio-therapy and the lack of published data regarding
the use of adjuvant chemotherapy, use of adjuvant chemotherapy is not justified
outside a clinical trial.
Prognosis : in most series, < 10% of
patients have local recurrence of disease and the 10-year disease free
and overall survival ranges from 50-80%, for the 30-50% of patients who
develop disease progression to myeloma. This occurs after a median of 1.5-2.5
years and their clinical course at progression is similar to those of patients
diagnosed with de novo symptomatic myeloma. Because of the small
number of patients in most series, any statistically significant risk factors
for development of myeloma are not clear and are further complicated by
the inclusion of patients over many decades during which treatment and
diagnostic modalities have become more refined and are likely to impact
prognosis. In some series, bulky disease > 5 cm may have prognostic significanceref.
In contrast to SBP, because < 25% of patients have a monoclonal protein,
any statistically significant analysis of disappearance of paraprotein
is precluded for patients with EMP
-
indolent myeloma : a variant of MM
in which the tumor cells are hypoproliferative; an M component and bone
marrow plasmacytosis are present, but significant bone marrow destruction,
hypercalcemia, and Bence Jones proteinuria are absent. It is defined as
asymptomatic MM with < 4 osteolytic lesions and normal renal function.
MRI can detect involvement of the vertebral marrow in 50% of patientsref1,
ref2
-
plasma cell or plasmacytic leukemia
(PCL) (2-4% of all myelomasref1,
ref2,
ref3)
: a rare form of plasma cell dyscrasia characterized by Kyle's criteria
: the presence of > 20% plasma cells in peripheral blood and an absolute
plasma cell count > 2,000/ml (Grogan TM,
Muller-Hermelink HK, Van CB, Harris NL, Kyle RA. World Health Organization
Classification Tumours of Haematopoietic and Lymphoid Tissues. France:
IARC Press Lyon; 2001.pp. 142-56)
Epidemiology : in almost all the series
median age ranged between 53-57 years (about 10 years younger than median
age in MM series). The youngest age reported was 30 yearsref
PCL can be of 2 types :
-
primary PCL (PPCL) (60-80%) presents de
novo in patients with no previous history of MM and usually features
a rapid clinical progression and a short survival. PPCL is a distinct clinicopathological
entity, because its clinical features, response to chemotherapy and prognosis
are different from those of typical multiple myeloma. PPCL shows higher
expression of CD20 as compared to MMref.
Epidemiology : incidence < 1 case in
1 million
Symptoms & signs : less lytic bone
lesions (60%), hepatosplenomegaly and lymphadenopathy are more common;
PPCL has a more aggressive course high frequency of extramedullary involvement
(liver, spleen, lymph nodes, extra osseous plasmacytomas etc), thrombocytopenia,
anemia, hypercalcemia and impaired renal function
Laboratory examinations : in certain PCL
cases, the leukemic cells are more immature than usual and a definitive
diagnosis is difficult to obtain (small lymphoid or lymphoplasmacytoid
cells with numerous cytoplasmic hairy projections; CD19-20-38+49e-79a-;
cytoplasmic and urinary l-type Bence–Jones protein;
germinal center B-cellsref)
without electron microscopicref
or immunohistochemical studies.
Therapy : single alkylating agent with
prednisolone is not appropriate for patients with primary PCL. VAD
and EDAP
with rituximab
ref.
Survival is significantly better in PCL patients treated with polychemotherapy
as compared to melphalan and prednisolone. Drugs used are vincristine,
doxorubicin, dexamethasone and/or cyclophosphamide and etoposideref.
Alternatively VCMP
/VBAP
ref
or DMVM
ref
is also used. Since the prognosis is so poor, intensification of high dose
chemotherapy followed by allogenic/autologous hematopoietic
stem cell transplantation
should be triedref1,
ref2,
ref3,
ref4,
ref5
Prognosis : median survival is 6 monthsref.
The longest survival reported was 28 monthsref.
The failure to achieve 50% clearance of blood plasma cells within 10 days
after the initiation of treatment is a predictor of no responseref.
Garcia-Sanz et al.ref
identified 10 variables which have unfavorable prognostic value on the
survival of PPCL cases, of which serum b2
microglobulin level > 6 mg/L and S phase bone marrow plasma cells >4.5%
retained an independent value on multivariate analysis. Response to treatment
of PCL is poor.
-
secondary PCL (40%)ref
evolves as a terminal event in 1-2% of MMref;
it is mostly considered as a complication of the alkylating
agents
or radiation therapy
ref1,
ref2
Epidemiology : older than PPCL
Symptoms & signs : hepatosplenomegaly
and lymphadenopathy are less common; more lytic bone lesions (100%), lower
platelet count, larger M-protein in the serum than did patients with primary
plasma cell leukemia
Prognosis : mean survival is 1.3ref-2.0
monthsref
Therapy : bortezomib
ref
Pathogenesis : deletions of 13q14 were detected
in 11 (78%) cases and deletions of 17p13.1(TP53) in 6 (43%) cases. Translocations
(11;14), (4;14), and (14;16) were found in 5 (35%), 2 (14%), and 1(7%)
cases, respectively. Except for an association between t(4;14) and 13q14
deletions, no association was identified among the genetic abnormalitiesref.
Phenotypically they originate from proliferation of plasma cells expressing
CD38ref.
Minority of cells expresses CD10, HLA-DR and CD20. The presence of multiple
haemopoietic surface antigens on malignant plasma cells suggests its origin
from a pluripotent stem cell. Also, plasma cells from both primary and
secondary PCL lacks CD56, which is important in anchoring plasma cells
to bone marrow stroma. > 80% have a diploid/hypodiploid DNA contentref.
Cytogenetic study shows complex karyotype with multiple numerical and structural
abnormalitiesref.
Up to 90% may show chromosome 13 monosomyref.
Among immunoglobulins, IgG is most often increased.
Laboratory examinations : anaemia with a haemoglobin
< 9 g/dl occurs in 80% of cases of PCL versus 35% of cases of MM. Rouleaux
formation is usually evident on the peripheral blood smear. Thrombocytopenia
with platelets < 100 x 109/L occurs in 50% of patients with
PCL versus only 10% of those with MM. Leucocytosis ranges from 20 to >
100 x 109/L with 20% to 100% of plasma cells. An elevated BUN
and/or creatinine occur in 75% of cases of PCL versus only 40% cases MM.
Most patients with PCL have a monoclonal IgG heavy chain or light chain
in the serum and Bence Jones proteinuria occurs in about 80% of casesref
Therapy : arsenic
trioxide (ATO)
.
SPCL rarely responds to chemotherapy.infection and haemorrhage contribute
significantly to morbidity and mortality. DIC has also been reportedref.
Differential diagnosis : acalculous cholecystitisref
-
‘hairy-cell’ variant of plasma cell myeloma (Crogan TM, Spier CM.
B-cell Immunoproliferative disorders, including multiple myeloma and amyloidosis.
In: Knowles DM, editor. Neoplastic hematopathology, 2nd ed. Philadelphia:
Lippincot Williams and Wilkins 2001;1564–5)
-
lymphoplasmacytoid leukemia producing biclonal IgA but not Bence–Jones
proteinref
Durie-Salmon and Bataille stagingref1,
ref2
:
-
I (approx. cell mass : 0.6 x 1012/m2; <
500 cells / mL; low) : all of the following
-
hemoglobin > 10 g/dl
-
serum calcium < 12 mg/dl (normal)
-
on roentgenogram, normal bone structure (scale 0) or solitary bone plasmacytoma
only
-
low M component production rates
-
IgG value < 5 g/dl
-
IgA value < 3 g/dl
-
urine light chain M-component on electrophoresis < 4 g/24h
-
II (approx.cell mass : 0.6-1.2 x 1012/m2;
500÷1200 cells / mL; intermediate) : overall data not as minimally
abnormal as shown for stage I and no single value as abnormal as defined
for stage III
-
III (approx.cell mass > 1.2 x 1012/m2; > 1200
cells/mL; high) : one or more of the following
-
hemoglobin value < 8.5 g/dl
-
serum calcium value > 12 mg/dl
-
advanced lytic bone lesions (scale 3)
-
high M-component production rates
-
IgG value > 7 g/dl
-
IgA value > 5 g/dl
-
urine light chain M-component on electrophoresis > 12 g/24 h
Subclassification :
-
A : relatively normal renal function (serum creatinine < 2.0 mg/dl)
-
B : abnormal renal function (serum creatinine > 2.0 mg/dl)
Symptoms & signs
:
-
multiple bone marrow tumor foci and secretion of an M component, associated
with widespread osteolytic lesions resulting in bone pain, pathologic
fractures
,
height decrease (=> spinal cord compression => paralysis)
-
hypercalcemia

-
normochromic normocytic anemia
-
spread to extraosseous sites occurs frequently in advanced disease
-
depression of immunoglobulin levels results in increased susceptibility
to opportunistic bacterial infections
-
renal failure
may also occur from .... :
These features have strong diagnostic value if accompanied by > 10% atypical
plasma cells in the bone marrow and either a monoclonal immunoglobulin
in the serum or light chains in the urine (Durie BGM, Salmon SE. Multiple
myeloma, macroglobulinaemia and monoclonal gammopathies. In: Hoffbrand
AV, Brain MC, Hirsh J, eds. Recent advances in haematology. No. 2. Edinburgh,
Scotland: Churchill Livingstone, 1977:243-61). Light chains appear in the
serum only if the patient has severe renal failure. Bone lesions, hypercalcemia,
and anemia correlate directly with the presence of the total mass of myeloma
cellsref
and have prognostic valueref1,
ref2,
ref3,
ref4.
All these complications, especially infection and renal insufficiency,
are also major causes of deathref.
The pathogenesis of these clinical features depends on interactions between
myeloma cells and the microenvironment of the bone marrow by means of cell-to-cell
contact, adhesion molecules, and cytokinesref
or on the direct effects of circulating monoclonal immunoglobulins or light
chainsref.
-
Crow-Fukase
syndrome / polyneuropathy, organomegaly, endocrinopathy, monoclonal gammapathy
and skin changes (POEMS) syndrome

-
sclerosing myeloma : myeloma associated
with osteosclerosis
,
most often manifested by peripheral
neuropathy
;
the myeloma involved may be localized or a part of MM; POEMS syndrome may
be present
Laboratory
examinations :
-
serum protein electrophoresis (SPE) and immunofixation with % of eventual
monoclonal component(s) in g-globulins
-
IgG MM (60%)
-
IgA MM (20%)ref
-
IgE MMref
(0.01% in all plasmocytomasref)
: < 40 cases of IgE myeloma have been describedref1,
ref2,
ref3,
ref4,
ref5,
ref6,
ref7,
ref8,
ref9,
ref10,
ref11,
ref12,
ref13,
ref14;
more malignant clinical course; a case in which a monoclonal chain was
detected in the urine of the patient several years prior to diagnosisref;
associated with plasma cell leukaemiaref;
in a case an unusual picture was found in the urine where two IgE fragments,
namely an incomplete Fc fragment and an IgE specific Fab fragment were
excretedref
-
IgD MM (1-2% of all myelomas) : a small band
or no evident M-spike on serum electrophoresis and heavy light-chain proteinuria.
Thus, IgD myeloma can be considered a variant of Bence
Jones myeloma; the presence of the IgD M-protein and the predominance
of the l light chain are the only distinctive
features. Predominant occurrence in males and young patients. The median
survival of patients with IgD MM is almost 2 years, with 20% of them surviving
for > 5 years, frequent renal impairment, hypercalcemia and amyloidosis.
Outcome remains poor with chemotherapy and appears to be superior with
ASCTref.
Bence Jones myeloma should not be diagnosed without excluding the presence
of an IgD paraproteinref.
-
IgM MM
-
biclonal MM (0.1%)
-
urine protein immunofixation to detect Bence-Jones proteinuria (70% of
MM and 100% of micromolecular myeloma)
-
dosage of serum IgA, IgG, IgM
-
IgA
(n.v : 70-400 mg/dl)
-
IgG
(n.v. : 700-1600 mg/dl)
-
IgM
(n.v. : 40-280 mg/dl)
-
IgD
(n.v. 0.3-14 mg/dl)
-
IgE
(n.v. : < 0.025 mg/dl or < 150 E/l)
-
dosage of l and k
L chains in :
-
serum :
-
k (n.v. 1.7-3.7 g/l)
-
l (n.v. 0.9-2.1 g/l)
-
k/l ratio (n.v. 1.35-2.65)
-
urine :
-
k (n.v. : 0.0-7.1 mg/l)
-
l (n.v. : 0.0-3.9 mg/l)
-
hypogammaglobulinemia (93%) (Kyle RA, Greipp PR, Witzig TE, et al. An analysis
of 1027 newly diagnosed cases of multiple myeloma. Blood 1999;94:Suppl
1:537a-537a.abstract)
-
hypercalcemia
(30%) is uncommon at diagnosis
-
hypeuricemia (40%)
-
increased plasma viscosity (80%)
-
increase in Treg

-
increased [bone marrow AlkP
isozyme]plasma
-
increased [b2m
]plasma
-
CBC
-
increased ESR (95%), sideremia, transferrin, ferritin, bilirubin, SGOT,
SGPT,
g-GT, alkaline phosphatase, LDH, fibrinogen,
total serum proteins; IL-6, IL-6R, TDK
-
at diagnosis : mosaic HAV, HBV, HCV antigens/serology
-
skull X-rays
: rounded "punched out" lesions

-
bone
marrow trephine biopsy (BMTB)
:
-
myeloma cell : found in bone marrow and occasionally in peripheral
blood
-
in the more anaplastic forms, the cell is large, has abundant blue-staining
cytoplasm with no perinuclear pallor, and has one or more moderately large
and vesicular nuclei that may be centrally or eccentrically placed and
may contain nucleoli
-
in better differentiated tumors, the cell is smaller and, except for the
finer chromatic structure, greatly resembles a plasmacyte
-
Mott cell / berry, grape, or morular cell
(an abnormal plasma cell that contains Mott bodies or Russell
bodies
)
-
immunophenotype : CD19+/-209%28+33+/-38++43+/-459%525%54
/ ICAM-1+5672%117+(18%)138
/ syndecan 1+227
/ EMA+/-, HM1.24+, SP17+,
MUC1+,
sIg-,
cIghi. (i) a universal marker of both normal and malignant
plasma cells, CD138;
(ii) markers related to malignancy i.e., CD19,
CD27, CD28,
and CD56;
(iii) markers associated with signaling and severity of MM (CD45,
CD221). Different entities of MM can be defined on the basis of expression
of CD19,
CD20,
CD27 and CD117ref
-
12-30% of MM patients have coexistent AL
amyloidosis
ref
Durie criteria, 1986 :
-
major criteria :
-
histological diagnosis of plasmacytoma at bone or soft tissue biopsy
-
bone marrow plasma cells > 20%
-
M component
-
IgG > 3.5 g/dl
-
IgA > 2 g/dl
-
Bence-Jones proteinuria > 1g/24h
-
urinary k or l chain
> 1 g/24hr
-
minor criteria
-
bone marrow plasma cell 10-20%
-
M component
-
IgG < 3.5 g/dl
-
IgA < 2 g/dl
-
Bence-Jones proteinuria < 1 g/24h
-
osteolytic lesions
-
suppression of normal IgG (IgG < 600 mg/dl, IgA < 100 mg/dl, IgM
< 50 mg/dl)
International Myeloma Working Group diagnostic criteria for MM requiring
systemic therapy, 2006 : 1 major + 1 minor criteria OR 3 minor criteria
-
presence of an M-component (in patients with no detectable M-component,
an abnormal serum FLC ratio on the serum FLC assay can substitute and satisfy
this criterion. For patients, with no serum or urine M-component and normal
serum FLC ratio, the baseline bone marrow must have 10% clonal plasma cells;
these patients are referred to as having 'non-secretory myeloma'. Patients
with biopsy-proven amyloidosis and/or systemic light chain deposition disease
(LCDD) should be classified as 'myeloma with documented amyloidosis' or
'myeloma with documented LCDD,' respectively if they have >= 30% plasma
cells and/or myeloma-related bone disease) in serum and/or urine plus clonal
plasma cells in the bone marrow and/or a documented clonal plasmacytoma
-
PLUS one or more of the following related organ or tissue impairment
(ROTI) (end-organ damage according to CRAB criteria)(must be
attributable to the underlying plasma cell disorder)
-
Calcium elevation (>11.5 mg/dl) [>2.65 mmol/l]
-
Renal insufficiency (creatinine >2 mg/dl) [177 mmol/l
or more]
-
Anemia (hemoglobin <10 g/dl or 2 g/dl <normal) (hemoglobin
<12.5 mmol/l (hemoglobin of 10 g/dl is 12.5 mmol/l) or 1.25 mmol/l<normal)
-
Bone disease (lytic lesions or osteopenia)
International Myeloma Working Group uniform response criteria (all response
categories require two consecutive assessments made at anytime before the
institution of any new therapy; complete and PR and SD categories also
require no known evidence of progressive or new bone lesions if radiographic
studies were performed. Radiographic studies are not required to satisfy
these response requirements.)
-
complete response (CR) :
-
Negative immunofixation on the serum and urine and
-
Disappearance of any soft tissue plasmacytomas and
-
5% plasma cells in bone marrow (confirmation with repeat bone
marrow biopsy not needed)
-
stringent complete response (sCR) :
-
CR as defined above plus
-
Normal FLC ratio and
-
Absence of clonal cells in bone marrowb by immunohistochemistry
or immunofluorescence (presence/absence of clonal cells is based upon the
k/l ratio. An abnormal k/l ratio by immunohistochemistry and/or immunofluorescence
requires a minimum of 100 plasma cells for analysis. An abnormal ratio
reflecting presence of an abnormal clone is k/l of >4:1 or <1;2. Alternatively,
the absence of clonal plasma cells can be defined based on the investigation
of phenotypically aberrant PC. The sensitivity level is 10-3 (less than
one phenotypically aberrant PC within a total of 1000 Pc). Examples of
aberrant phenotypes include (1) CD38 +dim and CD56+ strong and CD19- and
CD45-; (2) CD38+dim and CD138+ and CD56++ and CD28+; (3) CD138+, CD19-
CD56++, CD117+)
-
very good partial response (VGPR) :
-
serum and urine M-component detectable by immunofixation but not on electrophoresis
or
-
>= 90 reduction in serum M-component plus urine M-component <100 mg
per 24 h
-
partial response (PR) :
-
50% reduction of serum M-protein and reduction in 24-h urinary M-protein
by 90% or to <200 mg per 24 h
-
if the serum and urine M-protein are unmeasurable (Refer to Table 4 for
definitions of measurable disease) a 50% decrease in the difference between
involved and uninvolved FLC levels is required in place of the M-protein
criteria2, 3
-
if serum and urine M-protein are unmeasurable, and serum free light assay
is also unmeasurable, 50% reduction in plasma cells is required in place
of M-protein, provided baseline bone marrow plasma cell percentage was
30%
-
in addition to the above listed criteria, if present at baseline, a 50%
reduction in the size of soft tissue plasmacytomas is also required
-
stable disease (SD) (not recommended for use as an indicator of
response; stability of disease is best described by providing the time
to progression estimates) Not meeting criteria for CR, VGPR, PR or
progressive disease
1. Wei A, Juneja A. Bone marrow immunohistology of plasma cell neoplasms.
J Clin Pathol 2003; 56: 406–411.
2. San Miguel JF, Almeida J, Mateo G et al. Immunophenotypic
evaluation of the plasma cell compartment in MM: a tool for comparing the
efficacy of different treatment strategies and predicting outcome. Blood
2002; 99: 1853–1856.
-
progressive disease (to be used for calculation of time to progression
and progression-free survival end points for all patients including those
in CR (includes primary progressive disease and disease progression
on or off therapy) requires any one or more of the following:
-
increase of 25% from baseline in :
-
serum M-component and/or (the absolute increase must be 0.5 g/dl) (for
progressive disease, serum M-component increases of 1 gm/dl are sufficient
to define relapse if starting M-component is 5 g/dl)
-
urine M-component and/or (the absolute increase must be 200 mg/24 h
-
only in patients without measurable serum and urine M-protein levels: the
difference between involved and uninvolved FLC levels. The absolute increase
must be >10 mg/dl.
-
bone marrow plasma cell percentage: the absolute % must be 10%c
-
definite development of new bone lesions or soft tissue plasmacytomas or
definite increase in the size of existing bone lesions or soft tissue plasmacytomas
-
development of hypercalcemia (corrected serum calcium >11.5 mg/dl or 2.65
mmol/l) that can be attributed solely to the plasma cell proliferative
disorder
-
clinical relapse requires one or more of:
-
direct indicators of increasing disease and/or end organ dysfunction (CRAB
features)b It is not used in calculation of time to progression or progression-free
survival but is listed here as as something that can be reported optionally
or for use in clinical practice
-
development of new soft tissue plasmacytomas or bone lesions
-
definite increase in the size of existing plasmacytomas or bone lesions.
A definite increase is defined as a 50% (and at least 1 cm) increase as
measured serially by the sum of the products of the cross-diameters of
the measurable lesion
-
hypercalcemia (>11.5 mg/dl) [2.65 mmol/l]
-
decrease in hemoglobin of 2 g/dl [1.25 mmol/l] (see Table 3 for further
details)
-
rise in serum creatinine by 2 mg/dl or more [177 mol/l or more]
-
relapse from CR (to be used only if the end point studied is DFS)
(for purposes of calculating time to progression and progression-free survival,
CR patients should also be evaluated using criteria listed above for progressive
disease) : any one or more of the following:
-
reappearance of serum or urine M-protein by immunofixation or electrophoresis
-
development of 5% plasma cells in the bone marrow (relapse from CR has
the 5% cutoff versus 10% for other categories of relapse)
-
appearance of any other sign of progression (i.e., new plasmacytoma, lytic
bone lesion, or hypercalcemia see below)
Practical details of response evaluation :
-
laboratory tests for measurement of M-protein
-
serum M-protein level is quantitated using densitometry on serum protein
electrophoresis (SPEP) except in cases where the SPEP is felt to be unreliable
such as in patients with IgA monoclonal proteins migrating in the beta
region. If SPEP is not available or felt to be unreliable (e.g., in some
cases of IgA myeloma) for routine M-protein quantitation during therapy,
then quantitative immunoglobulin levels on nephelometry or turbidometry
can be accepted. However, this must be explicitly reported, and only nephelometry
can be used for that patient to assess response and SPEP and nephelometric
values cannot be used interchangeably.
-
urine M-protein measurement is estimated using 24-h urine protein electrophoresis
(UPEP) only. Random or 24 h urine tests measuring kappa and lambda light
chain levels are not reliable and are not recommended
-
definitions of measurable disease :
-
response criteria for all categories and subcategories of response except
CR are applicable only to patients who have 'measurable' disease defined
by at least one of the following three measurements:
-
serum M-protein 1 g/dl (10 gm/l)[10 g/l]
-
urine M-protein 200 mg/24 h
-
serum FLC assay: Involved FLC level 10 mg/dl (100 mg/l)
-
response criteria for CR are applicable for patients who have abnormalities
on one of the three measurements. Note that patients who do not meet any
of the criteria for measurable disease as listed above can only be assessed
for stringent CR, and cannot be assessed for any of the other response
categories
-
follow-up to meet criteria for PR or SD
-
it is recommended that patients undergoing therapy be tracked monthly for
the first year of new therapy and every other month thereafter
-
patients with 'measurable disease' as defined above need to be followed
by both SPEP and UPEP for response assessment and categorization
-
except for assessment of CR, patients with measurable disease restricted
to the SPEP will need to be followed only by SPEP; correspondingly, patients
with measurable disease restricted to the UPEP will need to be followed
only by UPEP
-
patients with measurable disease in either SPEP or UPEP or both will be
assessed for response only based on these two tests and not by the FLC
assay. FLC response criteria are only applicable to patients without measurable
disease in the serum or urine, and to fulfill the requirements of the category
of stringent CR
-
to be considered CR, both serum and urine immunofixation must be carried
out and be negative regardless of the size of baseline M-protein in the
serum or urine; patients with negative UPEP values pretreatment still require
UPEP testing to confirm CR and exclude light chain or Bence–Jones escape
-
skeletal survey is not required for assessment of response unless clinically
indicated, but is recommended once a year in clinical practice; bone marrow
is required only for categorization of CR, and for patients with non-secretory
disease
Therapyref
: IMiDsref
and bortezomibref
have been shown to inhibit human MM cell growth, decrease tumor-associated
angiogenesis, and prolong host survival in models of human MM xenografts
implanted subcutaneously in SCID mice. Subsequent Phase I, II and III clinical
trials of IMiDsref
and bortezomibref
have already demonstrated marked clinical activity even in patients with
refractory and relapsed MM. Corresponding in vitro gene array studies
of MM tumor cells before and after treatment with novel anti-MM agentsref1,
ref2,
ref3,
ref2
have helped to identify in vivo targets and mechanisms of novel
drug action as well as mechanisms of drug resistance. This helps to determine
whether in vivo targets of these novel therapies correlate with
their in vitro anti-MM activities in relapsed and refractory disease.
Extensive studies have focused on the mechanisms whereby MM cells home
to the host BM and adhere to BM stromal cells (BMSCs) and ECM proteins,
and the functional sequelae of these interactions. Such studies have identified
a series of cell adhesion molecules mediating MM cell binding to fibronectin
and BMSCs, thereby increasing MM cell proliferation and survivalref.
This proliferative and anti-apoptotic advantage conferred by stromal adhesion
is largely due to BMSC-derived cytokines, such as IL-6ref
and IGF-1ref1,
ref2.
In addition, the MM cell-BMSC binding interaction modulates the expression
of other cytokines, such as VEGF, which in turn stimulates increased local
angiogenesisref,
thereby further facilitating MM cell proliferation and viability. Importantly,
these aforementioned cytokines/growth factors can cooperate, in additive
and/or synergistic manners, to stimulate MM cell proliferation and survivalref1,
ref2,
ref3.
Antitumor activity of novel agents in animal models : the SCID-hu model
of human MM has been developed to allow studies of the dynamic interaction
between MM cells and the BM stroma, and to examine the efficacy of novel
therapies in that setting. Human bone grafts are implanted bilaterally
in the flanks of SCID mice. Human MM cells implanted into these grafts
proliferate, secrete MM idiotypic protein detectable in mouse serum, and
can migrate predominantly to the contralateral human BM graftref.
This in vivo model of human MM has provided a means to evaluate
the mechanisms mediating the specific homing of human MM cells to the human
BM microenvironment, as opposed to the murine BM microenvironment, as well
as for studying the role of microenvironmental factors (host-MM cell interactions,
cytokines, angiogenesis) in MM pathogenesis. In vivo assays of drug
efficacy have also used a beige-nude-xid mouse model of subcutaneous human
plasmacytoma xenografts. These models were critical for preclinical evaluation
of Thal, IMiDs, bortezomib and other novel agents. Clinical Trials of Agents
Targeting MM and the BM Microenvironment
-
induction therapy :
-
first line :
-
IFN-a2b
has been shown to be effective in about 20% of patients
-
chemotherapy : in most patients, the fraction of proliferating cells is
< 1% until late in the diseaseref.
-
melphalan
+ prednisone
(MP) was the standard first-line treatment for elderly MM patients
ineligible for stem cell transplant since 1960. However, complete responses
(CRs) are rare
-
cyclophosphamide
+ prednisone
-
thalidomide

-
alone or thalidomide
+ dexamethasone : 28 patients with previously untreated asymptomatic
MM were treated with thalidomide 100 to 200 mg orally (PO) at bedtime (qhs)
with serial increments of 50 to 100 mg at weekly intervals, as tolerated
to a maximum of 600 mg PO qhs. 40 consecutive previously untreated patients
with symptomatic MM were also treated as above (maximum dose 400 mg) and
received dexamethasone 20 mg/m2 for 4 days beginning on days
1, 9, and 17; the second and third cycles of repeated dexamethasone were
begun on day 30. Both groups of patients were treated for at least 3 months.
The response rate was 36% for patients treated with thalidomide alone and
72% for patients treated with Thal-Dex, the latter including complete remission
in 16% of patients. The median time to remission was 4.2 months with thalidomide
alone and 0.7 months with thalidomide-dexamethasone. Grade 3 toxicity included
infections (9 patients) and thrombotic/embolic events (7 patients). 5 deaths
have occurred as a result of MM (2 patients), infection (1 patient), unknown
cause (1 patient), and a possible thromboembolic event (1 patient)ref
-
thalidomide
+ dexamethasone
(Thal-Dex) : administered for 75 days (200 mg/day) and 3 months, respectively.
The monthly dose of dexamethasone was 20 mg/m2/day for 4 days,
with cycles repeated on days 9 to 12 and 17 to 20 on the first and the
third month of therapy.
-
is better than dexamethasone alone : the response rate with thalidomide
200 mg per day + dexamethasone 40 mg was significantly higher than with
dexamethasone 40 mg alone (63% v 41%, respectively) in patients with newly
diagnosed myeloma for 4 cycles (28 days per cycle), followed by stem cell
collection in patients eligible for high-dose chemotherapy (ECOG-EA100).
A recent interim analysis of 109 patients showed an overall response rate
of 80% ( 50% reduction in serum and urine M-protein) in the Thal/Dex arm,
significantly higher than 53% for patients on Dex alone (P = .0023; one-sided
Fisher's exact test). The most important side effect reported was deep
vein thrombosis (including pulmonary embolus), occurring in 16% of patients
receiving the Dex/Thal combination, compared with 3% receiving Dex alone
(Rajkumar SV, Blood E, Vesole DH, Shepard R, Greipp PR. A randomised phase
III trial of thalidomide plus dexamethasone versus dexamethasone in newly
diagnosed multiple myeloma (E1A00): a trial coordinated by the Eastern
Cooperative Oncology Group. Proc Am Soc Clin Oncol; 2004:558). The response
rate allowing for use of serum monoclonal protein levels when a measurable
urine monoclonal protein was unavailable at follow-up was 72% v 50%, respectively.
The incidence rates of grade 3 or higher deep vein thrombosis (DVT), rash,
bradycardia, neuropathy, and any grade 4 to 5 toxicity in the first 4 months
were significantly higher with thalidomide plus dexamethasone compared
with dexamethasone alone (45% v 21%, respectively; P < .001). DVT was
more frequent in arm A than in arm B (17% v 3%); grade 3 or higher peripheral
neuropathy was also more frequent (7% v 4%, respectively)ref
-
is better than VAD, Thal-Dex resulted in a significantly higher
response rate (52% versus 76%, respectively; P < .001) and effected
more profound reduction in myeloma cell mass of both IgG and IgA type.
More frequent toxicities included nonfatal deep vein thrombosis with Thal-Dex
(15%) and granulocytopenia with VAD (12%). In each of the 2 treatment groups,
91% of patients proceeded to PBSC mobilization. The median number of collected
CD34+ cells was 7.85 x 106/kg in the Thal-Dex group
and 10.5 x 106/kg in the control group. Thal-Dex may be considered
an effective and relatively well-tolerated oral alternative to the more
complex VAD regimen as front-line therapy for MM patients who are candidates
for subsequent autologous transplantationref
-
in young patients (< 61 years) with symptomatic MM, on an ITT basis,
the overall response (> or = PR) rate was 74%, including 24% of patients
who obtained a complete remission. Grade 3-4 toxicities consisted of infections
(12%), deep-vein thrombosis (3%), constipation (5%), and neuropathy (5%).
A total of 58 patients (96%) proceeded to PBSC mobilisation and yielded
a median number of 8 x 106 CD34+ cells/kg.ref.
-
In the Mayo Clinic Phase II trial, patients received Thal 200 mg per day
and Dex 40 mg on days 1-4, 9-12, and 17-20 (odd cycles) and 40 mg/d on
days 1-4 (even cycles); a 64% rate of major response, defined as
50% reduction in serum and urine M-protein, was reported. The most common
grade 3/4 toxicities were deep vein thrombosis (12%), constipation (8%),
rash (6%), and dyspnea (4%)ref.
-
The MD Anderson Cancer Center (MDACC) Phase II trial enrolled patients
with previously untreated symptomatic disease to receive Thal 100 mg per
day, escalated to 400 mg per day, plus Dex 20 mg. Partial and complete
responses were observed in 63% of patients, and grade 3 toxicities included
infection (23%), thrombotic/embolic events (15%), constipation (3%), and
fatigue (3%)ref.
-
thalidomide was given at the fixed dose of 200 mg/day; dexamethasone was
administered at the dose of 40 mg/day on days 1-4, 9-12 and 17- 20 in odd
cycles and 40 mg/day on days 1-4 in even cycles, repeated monthly for 4
months. 71 patients with symptomatic MM were evaluated for response and
toxicity. On an ITT basis, the overall response (>or= partial remission)
rate was 66%, including 17% of patients who attained a complete remission
or a very good partial remission. In addition to common toxicity of thalidomide,
deep-vein thrombosis was a troublesome adverse event (16%). 9 patients
(13%) required thalidomide discontinuation because of toxicity, including
3 patients who died during the study treatment. 59 patients proceeded to
PBSC mobilization and yielded a median number of 7.1x106 CD
34+ cells/kgref.
-
pegylated liposomal doxorubicin
(Caelyx) 40 mg/m2 on day 1 every 28 days, dexamethasone
40 mg p.o. on days 1-4 and 9-12 and thalidomide
100 mg daily (TAD / ThaDD)
-
in advanced MM achieved CR in 26%, nCR in 6%, VGPR in 6%, PR in 38%, minor
response in 16% and PD in 8% (ORR = 92%). The median EFS was 17 months
and the median OS was not reached. Grade 3 non-hematologic toxicity occurred
in 12% of patients, thromboembolic disease in 12% and severe infection
in 16%ref
-
phase II study in the treatment of 50 newly diagnosed MM patients > 65
years. Thalidomide was administered orally 100 mg at bedtime continuously,
dexamethasone orally 40 mg days 1-4 and 9-12 and pegylated liposomal doxorubicin
intravenously 40 mg/m2 on day 1 over the 28-days cycle. Response
was assessed according to the EBMT criteria. Seventeen (34%) patients achieved
CR, 7 (14%) nCR, 5 (10%) VGPR, 15 (30%) PR and 5 (10%) MR resulting in
an ORR of 98%. Only one patient (2%) presented progressive disease. TTP,
EFS and OS projected at 3 years were 60%, 57% and 74% respectively, and
these parameters were significantly higher in those patients achieving
a response VGPR versus those who did not. Grade 3-4 nonhematologic adverse
events were constipation (10%), fatigue (6%), tremors (4%), mucositis (4%)
and palmar-plantar erythro-dysesthesia (2%). Grade 3-4 neutropenia occurred
in 12% of patients. Grade 3-4 infections and thrombo-embolic accidents
were observed in 22% and 14% of patients, respectivelyref.
-
doxorubicin
and dexamethasone
were given for 2 or 3 months followed by thalidomide
and dexamethasone
for 2 months (AD-TD regimen) with prophylactic antibiotics and daily
aspirin (81 mg/d). Among the 42 patients whose response could be assessed,
38 responded to therapy (90.5%). The ITT response rate was 84.4% with 7
complete responses (CR 15.5%), 9 near complete responses (nCR 20.0%), and
22 partial responses (PR 48.9%). 2 patients had stable disease (4.4%),
and 2 progression of disease (4.4%). Normalization of the free light chain
ratio after 1 or 2 cycles of treatment was highly predictive of achievement
of CR or nCR. Patients tolerated the treatment well although 5 patients
developed thromboembolic complications (11%)ref
-
melphalan
+ prednisone
+
thalidomide
(MPT) : a multicentre randomised trial compared MP for 6 4-week cycles
plus thalidomide (100 mg per day continuously until any sign of relapse
or progressive disease) with MP alone in patients aged 60–85 years with
newly diagnosed MM. Patients treated with thalidomide had higher response
rates and longer event-free survival (primary endpoints) than patients
who were not. Combined CR or PR rates were 76.0% for MPT and 47.6% for
MP alone (absolute difference 28.3%), and the nCR or CR rates were 27.9%
and 7.2%, respectively. 2-year EFS rates were 54% for MPT and 27% for MP
(hazard ratio [HR] for MPT 0.51). 3-year OS rates were 80% for MPT and
64% for MP (HR for MPT 0.68). Rates of grade 3 or 4 adverse events were
48% in MPT patients and 25% in MP patients (p=0·.002). Introduction
of enoxaparin prophylaxis reduced rate of thromboembolism from 20% to 3%
(p=0·005)ref
-
BlT-D (Biaxin®
(clarithromycin)
500 mg twice daily, low-dose thalidomide
50-200 mg daily, and dexamethasone
40 mg weekly). Of the 50 patients available for analysis, 92% remain alive
and 64% remain on treatment with a median and mean duration of treatment
of 7 and 8 months, respectively. Overall, 93% of evaluable patients responded
to BLT-D, including 13% complete remissions, 40% near complete responses,
13% major responses, and 27% partial responsesref
The impact of thalidomide given in addition to chemotherapy throughout
the whole program is currently being tested in the Arkansas Total Therapy
II. When incorporated into high-dose, melphalan-based therapy for MM during
primary remission-induction therapy, between the 2 autologous HSCT, with
consolidation therapy, and as maintenance treatment, patients who received
thalidomide had a significantly higher rate of both complete response (62%
vs. 43%) and 5-year event-free survival (56% vs. 44%). Nevertheless, overall
survival curves in the 2 groups were nearly identical, owing in part to
the poorer outcome after relapse in the thalidomide group. In particular,
thalidomide-treated patients had a lower rate of response to salvage therapy
and shorter OS after relapse than the control patientsref.
These results indicate that contrary to a widely held belief, a CR is not
a valid surrogate for OS in clinical trials. The higher rate of failure
to respond to salvage therapy in the thalidomide group needs to be investigated
further, especially with respect to the salvage potential of new drugs
in patients who had received thalidomide as initial treatment.
-
lenalidomide
:
-
Revlimid + dexamethasone
(Rev-Dex) : 34 patients were enrolled. Lenalidomide was given orally
25 mg daily on days 1 to 21 of a 28-day cycle. Dexamethasone was given
orally 40 mg daily on days 1 to 4, 9 to 12, and 17 to 20 of each cycle.
Objective response was defined as a decrease in serum monoclonal protein
level by 50% or greater and a decrease in urine M protein level by at least
90% or to a level less than 200 mg/24 hours, confirmed by 2 consecutive
determinations at least 4 weeks apart. 21 of 34 patients achieved an objective
response, including 2 (6%) achieving complete response (CR) and 11 (32%)
meeting criteria for both very good partial response and near complete
response, resulting in an overall objective response rate of 91%. Of the
3 remaining patients not achieving an objective response, 2 had minor response
(MR) and one had stable disease. Fortyseven percent of patients experienced
grade III or higher nonhematologic toxicity, most commonly fatigue (15%),
muscle weakness (6%), anxiety (6%), pneumonitis (6%), and rash (6%). Rev/Dex
is a highly active regimen with manageable side effects in the treatment
of newly diagnosed MMref.
Rev-Dex is associated with an increased risk of venous thrombosis and concomitant
administration of erythropoietic agents may further increase the risk of
thrombosis (Dimopoulos M, Weber D, Chen C, et al. Evaluating oral lenalidomide
(Revlimid) and dexamethasone versus placebo and dexamethasone in patients
with relapsed or refractory multiple myeloma. Haematologica 2005;90:Suppl
2:160-160)ref.
Erythropoietin or other agents that may increase the risk of thrombosis
should be used with caution in patients receiving lenalidomide and high-dose
dexamethasone. Antithrombotic medications, including aspirin, should be
considered for such patients, especially in those with additional risk
factorsref.
-
BiRD (Biaxin®
(clarithromycin)
500 mg twice daily, lenalidomide
25 mg daily on days 1-21, and dexamethasone
40 mg weekly) in 28-day cycles. Objective response was defined by standard
criteria (ie, decrease in serum monoclonal protein [M-protein] by at least
50%, and a decrease in urine M-protein by at least 90%). Of the 72 patients
enrolled, 65 had an objective response (90.3%). A combined stringent and
conventional complete response rate of 38.9% was achieved, and 73.6% of
the patients achieved at least a 90% decrease in M-protein levels. This
regimen did not interfere with hematopoietic stem-cell harvest. 52 patients
who did not go on to receive transplants received continued therapy (complete
response, 37%; very good partial response, 33%)ref.
-
VAD (vincristine, adriamycin,
dexamethasone)
ref
followed by high-dose melphalan
with stem cell rescueref1,
ref2
-
pegylated liposomal
doxorubicin
+ vincristine
intravenous injection + oral dexamethasone
(DVD) is at least as effective as VAD/VAd for the treatment of MM,
but DVd is associated with less neutropenia and alopecia in addition to
requiring fewer days in the hospital or clinic for drug administration.
DVd therapy has also been reported to be associated with an antiangiogenic
effect not observed with VAD.
-
cyclophosphamide
,
vincristine
,
doxorubicin
and dexamethasone
(C-VAD)ref
-
bortezomib
is
currently being evaluated as part of induction treatment prior to ASCT,
alone or in combination with dexamethasone or with chemotherapy. It is
probably more attractive to test the impact of these new agents given in
addition to HDT rather than to again compare HDT with standard dose chemotherapy
including these drugs.
-
bortezomib
+ melphalan
+ prednisone
(VMP) : in a phase I/II trial in 60 untreated MM patients aged > 65
years (half >75 years), response rate was 89%, including 32% immunofixation-negative
CR, of whom half of those analyzed achieved immunophenotypic remission
(no detectable plasma cells at 10-4-10-5 sensitivity).
VMP appeared to overcome the poor prognosis conferred by Rb gene deletion
and IgH translocations. Results compare favorably with historical control
data for MP, notably response rate (89% vs 42%), EFS at 16 months (83%
vs 51%), and survival at 16 months (90% vs 62%). Side effects were predictable
and manageable; principal toxicities were hematologic, gastrointestinal,
and peripheral neuropathy, and were more evident during early cycles and
in patients aged 75 years. In conclusion, in elderly patients ineligible
for transplant, the combination of bortezomib plus MP appears significantly
superior to MP, producing very high CR rates, including immunophenotypic
CR, even in patients with poor prognostic featuresref
-
as second-line in relapsed, refractory MM :
-
high-dose dexamethasone
(40 mg orally on days 1 through 4, 9 through 12, and 17 through 20 for
4 5-week cycles)
-
thalidomide

-
alone (100-600 mg per day) : using response criteria defined as
>= 25% reduction in serum M-protein concentration, response rates are 30/70%,
with a median time to response ranging from 3 to 8 weeksref1,
ref2.
-
Thal + melphalan
and Thal + melphalan
+ dexamethasone
yielded response rates of 82% (> 25% reduction in M-protein) but significant
rates of myelotoxicity, with 87% and 62% of patients experiencing leukopenia
and neutropenia, respectivelyref1,
ref2
-
Thal + liposomal doxorubicin
+
vincristine
+ dexamethasone
(DVd-T) : an encouraging overall response rate of 74% was seen, but
an increased incidence of neutropenia, bacterial and viral infection, and
deep vein thrombosis was observed, prompting a protocol amendment to allow
concomitant treatments, which proved largely successful in abrogating these
toxicitiesref.
-
Thal +
cyclophosphamide
+ dexamethasone
(ThaCyDex) is effective in patients with
b2-microglobulin
</=4 mg/dL and </=65 yearsref
-
melphalan
50 mg/m2 and bortezomib
1.3 mg/m2 on days -6 and -3 in association with thalidomide
200 mg and dexamethasone
20 mg on days -6 through -3, followed by hematopoietic cell support on
day 0. Nonhematologic toxicities included pneumonia, febrile neutropenia,
and peripheral neuropathy. All patients had undergone autologous transplantation
at diagnosis, and 13 patients (50%) underwent an additional transplantation
at relapse. Responses occurred in 17 of 26 patients (65%), including 1
complete remission, 3 near complete remissions (12%), and 2 very good partial
remissions (8%). Response rate was higher than that induced by the previous
line of treatment in 12 patients (46%)ref
-
lenalidomide
ref
-
Rev alone :
-
phase I studies :
-
IMiD CC-5013 dose escalation (5 mg/day, 10 mg/day, 25 mg/day and 50 mg/day)
study in 27 patients (median age 57, range 40-71 years) who had received
a median of 3 (range 2-6) prior regimens, including autologous stem cell
transplantation and Thal in 15 and 16 patients, respectively. In 24 evaluable
patients, no dose-limiting toxicity was observed in patients treated at
any dose level within the first 28 days; however, grade 3 myelosuppression
developed after day 28 in all 13 patients treated with 50 mg/day CC-5013.
In 12 patients, dose reduction to 25 mg/day was well tolerated and therefore
considered the maximal tolerated dose. Importantly, no significant somnolence,
constipation, or neuropathy has been seen in any cohort. Best responses
of 25% reduction in paraprotein occurred in 17 of 24 (71%) patients
(90% confidence interval [CI]: 52%, 85%), including 11 (46%) patients who
had received prior Thal. Stable disease (< 25% reduction in paraprotein)
was observed in an additional 2 (8%) patients. Therefore, 17 of 24 (71%)
patients (90% CI: 52%, 85%) demonstrated benefit from treatmentref
-
Zangari M, Tricot G, Zeldis J, et al. Results of a phase I study of CC-5013
for the treatment of multiple myeloma (MM) patients who relapse after high
dose chemotherapy (HDCT) [abstract]. Blood. 2001:3226a
-
Revlimid + dexamethasone
(Rev-Dex) : Dimopoulos MA, Spencer A, Attal M, et al. Study of lenalidomide
plus dexamethasone versus dexamethasone alone in relapsed or refractory
multiple myeloma (MM): results of a phase 3 study (MM-010). Blood 2005;106:6a-6a
-
CC-4047
,
has been tested in a Phase I study of patients with relapsed MM, and results
have shown a favorable toxicity profile (although deep vein thrombosis
occurred in approximately 10% of patients) and high response ratesref
-
clarithromycin
is not effective as single-agent in advanced MMref1,
ref2, ref3,
but ...
-
clarithromycin
250 mg twice daily + thalidomide
50 mg at night on an ongoing basis + 4-d pulses of 10 mg dexamethasone
given monthly. 8 patients had permitted escalation of thalidomide dosage
up to 200 mg daily. The combination was well tolerated and could be given
to elderly, infirm and severely cytopenic patients. RRs were high, with
89% achieving at least 50% reduction in paraprotein and a 96% overall response
rateref
-
proteasome inhibitors
: are a novel class of therapeutics targeting the MM cell in its BM microenvironment
by blocking NF-kB-mediated IL-6 production.
Bortezomib
has been shown to induce apoptosis of MM cells resistant to known conventional
therapies. It overcomes the protective effects of IL-6, and adds to the
anti-MM effects of Dexref.
It acts in the BM microenvironment to inhibit the binding of MM cells to
BMSCs, the transcription and secretion of IL-6 triggered by MM to BMSC
adhesion, and BM angiogenesis. Gene microarray profiling studies demonstrate
that bortezomib induces transcriptional downregulation of growth/survival
signaling pathways and upregulates apoptotic cascades, ubiquitin/proteasome
pathways, and heat-shock proteinsref.
As expected, mean maximum plasma concentration was lower and took longer
to reach following subcutaneous administration. Overall 20S proteasome
inhibition was similar between arms. Safety profile and response rate for
the subcutaneous arm did not appear inferior to the intravenous arm, with
good local tolerance of subcutaneous injectionref.
-
alone (1.3 mg/m2BSA) on days 1, 4, 8, and
11 for 8 3-week cycles, followed by treatment on days 1, 8, 15, and 22
for 3 5-week cycles) : in a multicenter Phase II trial of bortezomib in
patients with advanced and heavily pretreated MM, CR was seen by Blade
criteria in 10% of patients, with 4% immunofixation negative and 6% with
residual MM paraprotein detected only by immunofixationref.
The overall response rate (MR + PR + CR) was 35% with an additional 24%
experiencing stable disease. In a recently updated analysis of time-to-event
parameters, median TTP was 7 months and median duration of survival was
17 months, with the median duration of response 12.7 months in those patients
with PR and CRref.
Responses were associated with improved hemoglobin levels, decreased blood
transfusion requirements, improvement in renal function and normalization
of uninvolved immunoglobulins. Drug-related gastrointestinal toxicity and
fatigue were in most cases manageable; significant thrombocytopenia and
neuropathy occurred primarily in patients in whom these conditions were
pre-existent, with treatment-emergent neuropathy seen in about a third
of patients, and serious adverse events were relatively uncommonref.
-
is superior to high-dose dexamethasone
(40 mg orally on days 1 through 4, 9 through 12, and 17 through 20 for
4 5-week cycles) for the treatment of patients with MM who have had a relapse
after 1 to 3 previous therapiesref.
Response rates were superior to those observed with dexamethasone regardless
of the type of first-line therapy, with the exception of prior thalidomide
treatment (Sonneveld P, Richardson PG, Schuster MW, et al. Bortezomib at
first relapse is superior to high-dose dexamethasone and more effective
than when given later in relapsed multiple myeloma. Haematologica 2005;90:Suppl
1:146-146). Based upon preclinical studies showing an additive anti-MM
activity of Dex and bortezomibref,
Dex was added in those patients who either progressed or achieved only
stabilization of disease on bortezomib alone, with additional responses
seen in 18% of those evaluableref.
As a result of these encouraging data, US Food and Drug Administration
(FDA) approval for bortezomib in the treatment of relapsed and refractory
MM was given in 2003. An interim analysis of a Phase III international,
randomized trial of bortezomib versus high-dose Dex in relapsed MM showed
significant benefit in the bortezomib arm as assessed by time to progression
and OS, resulting in the closure of the Dex arm and permitting all patients
to cross over to bortezomib (Richardson PG, Sonneveld P, Schuster MW, et
al. Bortezomib vs. dexamethasone in relapsed multiple myeloma: a phase
3 randomized study. Proc Am Soc Clin Oncol. 2004;23:558).
According to Trippoli et al.ref
and Sonneveld et al.ref,
life expectancy for patients with myeloma who are alive at 1 year after
treatment is about 1.5 additional years (range, 0.253 to 2.472). Bortezomib
costs € 49,077 ($59,991 in the USA at the current exchange rates)
per patient on the basis of the schedule outlined in the article and current
Italian prices. Considering that some treatment cycles are discontinued
early because of side effects or death, the incremental cost of bortezomib
as compared with dexamethasone is about € 40,000 ($48,942) per patient.
Improving survival by 14% at 1 year gives a lifetime gain of at least 21
years (14 x 1.5) for every 100 patients. On the basis of these data, the
cost per life-year gained for bortezomib as compared with dexamethasone
is € 190,476 ($232,924). This analysis suggests that the benefit of
bortezomib has a cost that exceeds current international benchmarksref.
Anyway the critic was partially faulty, since the mean number of treatment
cycles in the study was 6 rather than 11. The mean body-surface area of
patients was 1.89 m2 rather than 2 m2. Furthermore,
it is premature to use survival data to model cost-effectiveness on the
basis of data from the Assessment of Proteasome Inhibition for Extending
Remissions (APEX) trial, since fewer than one third of the events had occurred
at the cutoff point for data collection. In a comprehensive, published
analysis that was performed on the basis of data from patients with relapsed
and refractory myeloma and that accounted for the costs of therapy, disease
complications, and management of adverse events, bortezomib, as compared
with best supportive care, had an incremental cost-effectiveness ratio
of $45,356 per life-year gainedref.
In another analysis, bortezomib had an incremental cost-effectiveness ratio
as low as € 25,271 ($30,910) per life-year gained (Bagust A, Haycox
AR, Boland A, et al. Economic evaluation of bortezomib (VELCADE) for relapsed
and refractory multiple myeloma. Blood 2004;104:80a-80a. abstract), suggesting
that the ratio fell below the international benchmarkref1,
ref2.
These findings suggest that bortezomib is a cost-effective option for patients
with relapsed MMref
-
combination trials in which bortezomib has been administered with other
agents in patients otherwise resistant to each agent alone :
-
bortezomib
+ thalidomide
ref
-
bortezomib
+ thalidomide
+ dexamethasone
(VTD)ref1,
ref2,
ref3
-
Velcade (bortezomib)
,
Doxil
(pegylated liposomal doxorubicin)
and thalidomide
(VDT)ref
-
bortezomib
+ melphalan
(Yang H, Swift R, Sadler K, et al. A phase I/II trial of Velcade and melphalan
combination therapy (Vc+M) for patients with relapsed or refractory multiple
myeloma (MM) [abstract]. Blood. 2003;102:235a)
-
bortezomib
+ dexamethasone
ref
-
bortezomib
+ pegylated liposomal
doxorubicin
ref1,
ref2
-
bortezomib
+ dexamethasone
+ doxorubicin
(Cavenagh JD, Curry N, Stec J, et al. PAD therapy (bortezomib, doxorubicin
and dexamethasone) for untreated multiple myeloma (MM) [abstract]. Proc
ASCO. 2004;23:568)
-
bortezomib
+ non-myeloablative 153Sm-EDTMP
skeletally targeted radiotherapy in an orthotopic model of MMref
-
Velcade©
(bortezomib)
,
melphalan
and prednisone
(VMP) demonstrated a CR(1) rate of 43%, the strongest rate ever reported
for a melphalan prednisone combination therapy. At 38 months, 85% of patients
were alive. This is the highest reported 3-year survival rate in the front-line
treatment settingref
-
Velcade©
(bortezomib)
,
adriamycin and dexamethasone
(referred to as VcAD or PAD) showed a CR(1) rate of 29 percent prior
to stem cell transplantation (SCT), which further improved to 57% following
SCT. At 1 year, 100% of patients were alive, and at 2 years, 95% of patients
were alive. This is the highest reported 2-year survival rate in the front-line
treatment setting. Median overall survival (OS) has not yet been reached
after 4 yearsref
-
Velcade©
(bortezomib)
,
Doxil©
(pegylated liposomal doxorubicin)
and dexamethasone
(VDD) showed a CR(1) rate of 43% prior to SCT, which increased to 65%
following SCT. At 16 months, 100% of patients were aliveref
-
Velcade©
(bortezomib)
,
Revlimid©
lenalidomide
and dexamethasone
(VRD) showed an overall response rate (ORR) of 100%, including a CR(1)
rate of 20%ref
-
arsenic trioxide (ATO)
ref
has been identified as a third agent targeting the MM cell interacting
with its BM microenvironment. As2O3 at clinically
achievable levels induces apoptosis of drug-resistant MM cell lines and
patient cells via caspase-9 activation, adds to Dex, and can overcome the
anti-apoptotic effects of IL-6ref.
It also decreases MM cell binding to BMSCs, inhibits IL-6 and VEGF secretion
in BMSCs induced by MM cell adhesion, and blocks proliferation even of
those MM cells adherent to BMSCs. Clinical trials of As2O3
are ongoing.
-
VEGFR inhibitors
(including PTK787/ZK222584) have been identified as a promising therapy
in MM as VEGF is expressed and secreted by MM cells and BM stromal cells
with VEGF secretion by MM cells augmenting IL-6 secretion in BM stromal
cellsref.
VEGF receptor tyrosine kinase inhibitor PTK787/ZK222584 blocks VEGF-induced
tyrosine phosphorylation of Flt-1, MEK/MAPK activation, and proliferation
as well as PKC activation-dependent migrationref.
These studies both define VEGF as a novel therapeutic target and provide
the basis for a current Phase II clinical trial of the oral agent PTK787/ZK222584
in patients with relapsed MM.
-
farnesyl transferase inhibitors
(FTI)
inhibit tumor cell growth both in vitro and in vivo. Cytokine
(IL-6, VEGF, IGF-1)-induced proliferation of MM cells is mediated, at least
in part, via the Ras/Raf/MAPK signaling, which provides the basis for both
ongoing and recently reported Phase I/II clinical trials of two promising
FTIs, SCH-66336 and R115777, in relapsed and refractory MMref
-
histone deacetylases (HDACs)
inhibitors
ref
: novel hydroxamic acid-based hybrid polar compounds, such as suberoylanilide
hydroxamic acid (SAHA), are histone deacetylase inhibitors that induce
differentiation and/or apoptosis selectively in transformed and neoplastic
cells. SAHA is an orally bioavailable HDAC inhibitor, which induces
in vitro growth arrest and apoptosis of primary tumor cells from MM
patients and cell lines, irrespective of their status of resistance to
other agentsref1,
ref2
-
+ bortezomib
ref.
Phase I studies in other malignancies have documented that SAHA is well
tolerated, and a Phase I trial of this promising novel agent for the treatment
of relapsed MM is underway. LBH589 is a novel hydroxamic acid derivative
that at low nanomolar concentrations induces apoptosis in MM cells resistant
to conventional therapies via caspase activation and poly (ADP-ribose)
polymerase (PARP) cleavage. Significant synergistic cytotoxicity was observed
with LBH589 in combination with bortezomib against MM cells which were
sensitive and resistant to Dexamethasone (Dex), as well as primary patient
MM cells. LBH589 at low nanomolar concentrations also induced -tubulin
hyperacetylation. Aggresome formation was observed in the presence of bortezomib,
and the combination of LBH589 plus bortezomib induced the formation of
abnormal bundles of hyeracetylated a-tubulin
but with diminished aggresome size and apoptotic nuclei. These data confirm
the potential clinical benefit of combining HDAC inhibitors with proteasome
inhibitors, and provide insight into the mechanisms of synergistic anti-MM
activity of bortezomib in combination with LBH589ref.
-
heat shock protein 90 (hsp90)
inhibitors
is a molecular chaperone that facilitates the intracellular trafficking,
conformational maturation, and proper 3-dimensional folding of a broad
range of intracellular proteins required for cell proliferation, survival
and drug resistance. Selective inhibitors of hsp90, such as ansamycin antibiotic
geldanamycin (GA) and its analogs (e.g., 17-allylamino-17-demethoxy-geldanamycin
or 17-AAG) have profound pro-apoptotic consequences on MM cells, regardless
of their resistance to other agents. Gene microarray profiling has shown
that bortezomib treatment induces a stress response in MM cells, which
includes the compensatory upregulation of hsp90, in an effort of the tumor
cells to escape the effect of proteasome inhibition; conversely, blocking
this protective stress response with GA or 17AAG enhances bortezomib-triggered
MM cell apoptosisref.
A Phase I study of single agent 17-AAG in relapsed and relapsed, refractory
MM patients is now ongoing (Mitsiades CS, Mitsiades N, Poulaki V, et al.
Hsp90 inhibitors prolong survival in a SCID/NOD mice model of diffuse multiple
myeloma: therapeutic implications. Blood. 2002;100:106a).
-
P38a MAPK inhibitors
mediates the production of various pro-inflammatory cytokines such as IL-1B,
IL-6 and TNFa, which play a critical role in
MM, and the targeting of P38a MAPK has been shown to inhibit MM cell growth
and survival in the BM milieuref.
An orally bioavailable P38a MAPK inhibitor (SCIO-469) is now entering Phase
II study in relapsed, refractory MM in combination with bortezomib.
-
IGF-1R / CD221 inhibitors
(NVP-ADW742)ref1,
ref2
-
PKB / Akt inhibitors
induces in vitro and in vivo cytotoxicity in human MM cellsref
-
2-methoxyestradiol (2ME2), a natural endogenous product
of estradiol metabolism, has demonstrated activity against tumor cell lines
and can inhibit angiogenesisref
-
PKC inhibitors
:
-
PKC412 induced apoptosis of HMCLs and primary MM cells with variable
efficacy, however, some activity was seen against all HMCLs and primary
MM cells with 0.5µM PKC412. PARP cleavage and decreased PKC activity
was observed in all HMCLs tested. Furthermore, PKC412 inhibited c-Fos transcription
and nuclear protein expression, induced ROS production and induced both
c-Jun expression and phosphorylation. The latter was inhibited by co-treatment
with the JNK inhibitor SP600125, which similarly abrogated PKC412-induced
apoptosis, suggesting that PKC412 induced apoptosis is a JNK dependent
event. PKC412 treatment secondarily induced pro-survival stress responses
as evidenced by activation of NFB and increased expression of the heat
shock proteins HSP70 and HSP90. Consistent with the former, sequential
inhibition of NFB activation with bortezomib or SN50 synergistically enhanced
cell killingref
-
enzastaurin is an oral drug that specifically inhibits phorbolester-induced
activation of PKC isoforms, as well as phosphorylation of downstream signaling
molecules MARCKS and PKCµ. Importantly, it also inhibits PKC-activation
triggered by growth factors and cytokines secreted by bone marrow stromal
cells (BMSCs); co-stimulation with fibronectin, VEGF or IL-6; as well as
MM patient serum. Consequently, enzastaurin inhibits proliferation, survival
and migration of MM cell lines and MM cells isolated from multidrug-resistant
patients; as well as overcomes MM cell growth triggered by binding to BMSCs
and endothelial cells. Importantly, strong synergistic cytotoxicity is
observed when enzastaurin is combined with bortezomib, moderate synergistic
or additive effects when combined with melphalan or lenalidomide. Finally,
tumor growth, survival and angiogenesis are abrogated by enzastaurin in
an in vivo xenograft model of human MMref.
-
a low molecular weight Smac mimetic LBW242 induces apoptosis in
MM cells resistant to conventional therapies and bortezomib. Examination
of purified patient MM cells demonstrated similar results, without significant
cytotoxicity against normal lymphocytes and bone marrow stromal (BMSCs)
cells. Importantly, LBW242 abrogates paracrine MM cell growth triggered
by their adherence to BMSCs and overcomes MM cell growth and drug-resistance
conferred by interleukin-6 or insulin-like growth factor-1. Overexpression
of Bcl-2 similarly does not affect LBW242-induced cytotoxicity. Mechanistic
studies show that LBW242-induced apoptosis in MM cells is associated with
activation of caspase-8, caspase-9 and caspase-3, followed by PARP cleavage.
In human MM xenograft mouse models, LBW242 is well tolerated, inhibits
tumor growth, and prolongs survival. Importantly, combining LBW242 with
novel agents including TRAIL or proteasome inhibitors bortezomib and NPI-0052;
as well as with conventional anti-MM agents melphalan or dexamethasone
induces additive/synergistic anti-MM activityref
-
radiotherapy
: MM cells are highly radiosensitive, and local radiotherapy provides effective
palliation for painful bone lesionsref.
However, the disseminated nature of MM precludes curative external beam
radiation therapy due to unacceptable end organ toxicityref.
Bone-seeking radioisotopes that bind to bone mineral are being tested in
MMref,
but their appeal and efficacy are limited by their inability to penetrate
into the centers of myelomatous bone marrow deposits.
-
(remission) maintenance therapy :
-
thalidomide
: preliminary results of the IFM99-02 trial show that, in patients with
standard prognosis, maintenance treatment with thalidomide significantly
prolongs EFS, as compared with no maintenance therapyref.
It will be important to assess whether relapses of aggressive myeloma,
such as those reported by Barlogie et al., also occur when thalidomide
is reserved for maintenance therapyref
or combined initially with chemotherapy at conventional doses (Palumbo
A, Bringhen S, Musto P, et al. Oral melphalan, prednisone and thalidomide
for multiple myeloma. Blood 2005;106:230a-230a). Another matter of importance
concerns the dose- and duration-related toxicity of thalidomide, a complication
that led to its discontinuation in 30% of patients within the first 2 years
and in > 60% within 4 years in the study by Barlogie and colleagues. As
in previous reports, deep-vein thrombosis and peripheral neuropathy were
the chief adverse effects of thalidomide therapy
-
prednisone
50 mg a.d.ref
-
IFN-a2b
3 MIU 2 times a week for (delays relapse or up to 10 years)
-
responding to conventional induction chemotherapyref
-
in first plateau phaseref1,
ref2
-
following high-dose chemotherapyref
-
after high-dose treatment and autologous HSCTref
-
IFN-a2b
+ prednisone
is more effective than IFN-a2b aloneref
:
-
hematopoietic
stem cell transplantation (HSCT)
ref1,
ref2,
ref3
:
-
autologous HSCT
: for patients responding to initial chemotherapy, ASCT is a safe (<
5% toxic deaths) and effective consolidation therapy. 30-50% complete remissions
(CR) could be achieved with this approach in newly diagnosed MM and this
more important tumor burden reduction can be converted into a prolongation
of remission and of survival. Thanks to autologous transplantation of peripheral
blood progenitor cells (PBPC) and to hematopoietic growth factors, the
sequential use of 2 courses of HDT has become more feasible and appears
to increase the CR rate and even to induce molecular remission), CD34+
selection, as well as modifications in the conditioning regimen to improve
outcomes. Sensitive PCR techniques using patient-specific
oligonucleotide primers show the persistence of up to 10% myeloma cells
in the CD34+
cell fractions while highly purified CD34+lin-90
/ THY1+ stem cells do no apparently contain clonal myeloma
cellsref1,
ref2,
but substantial delay of neutrophil and platelet engraftment compared to
unmanipulated PBPC grafts let the latter appear as the best source of stem
cell for ASCT in MM.
-
superiority of high-dose therapy (HDT) supported by autologous bone
marrow transplantation (BMT) compared to conventional chemotherapy (CC)
(alternating cycles of VMCP
and of BVAP
)
aministered at 3-week intervals for 12 months for a total of 18 cycles)
was first demonstrated by the Intergroupe Francophone du Myelome (IFM)
8 years agoref.
In this randomized IFM90 trial HDT significantly increased the complete
remission (CR) rate, the EFS and the OS in patients with newly diagnosed
MM up to the age of 65. Following this publication, the number of ASCT
performed worldwide as part of front-line therapy in younger patients increased
dramatically. However some concerns remained until last year when the IFM90
results were fully confirmed by a larger trial published by the British
Medical Research Council (MRC)ref.
Other randomized trials have been presented in meetings but not yet published
(Fermand JP, Ravaud P, Katsahian S, et al. High dose therapy and autologous
blood stem cell transplantation versus conventional treatment in multiple
myeloma: results of a randomized trial in 190 patients 55 to 65 years of
age [abstract]. Blood. 1999;94 (suppl 1):396a; Blade J, Sureda A, Ribera
JM, et al. High-dose therapy/autotransplantation/intensification versus
continued conventional chemotherapy in multiple myeloma patients responding
to initial chemotherapy. Definitive results from Pethema after a median
follow-up of 66 months [abstract]. Blood. 2003;102:42a; Palumbo A, Bringhen
S, Petrucci MT, et al. A prospective randomized trial of intermediate dose
melphalan (100 mg/m2) vs oral melphalan/prednisone [abstract].
Blood. 2003;102:984a; Barlogie B, Kyle R, Anderson K, et al. Comparable
survival in multiple myeloma with high dose therapy employing Mel 140mg/m2
+ TBI 12 Gy autotransplants versus standard dose therapy with VBMCP and
no benefit from interferon maintenance: results of Intergroup trial S9321
[abstract]. Blood. 2003;102:42a). Although the details of CC and of HDT
varied, the general outline of these studies were the same, except for
the Spanish group trial where only patients responding to their initial
CC were randomized.
study
|
no. of patients
|
age
|
CR Rate
(%)
(CC vs ASCT)
|
p value
|
median EFS
(months)
(CC vs ASCT)
|
p value
|
median OS
(months)
(CC vs ASCT)
|
p value
|
Intergroupe Francophone du Myelome (IFM90)ref |
200 |
< 65 |
5 vs 22 |
< 0.001 |
18 vs 28 |
0.01 |
44 vs 57 |
0.03 |
Medical Research Council (MRC7)ref |
401 |
< 64 |
8 vs 44 |
< 0.001 |
19 vs 31 |
< 0.001 |
42 vs 54 |
< 0.001 |
Myelome Auto Greffe (MAG91) (Fermand JP, Ravaud P, Katsahian S, et
al. High dose therapy and autologous blood stem cell transplantation versus
conventional treatment in multiple myeloma: results of a randomized trial
in 190 patients 55 to 65 years of age [abstract]. Blood. 1999;94 (suppl
1):396a) |
190 |
55-65 |
|
|
19 vs 25 |
0.05 |
45 vs 42 |
NS |
PETHEMA, Spanish Cooperative Group (Blade J, Sureda A, Ribera JM, et
al. High-dose therapy/autotransplantation/intensification versus continued
conventional chemotherapy in multiple myeloma patients responding to initial
chemotherapy. Definitive results from Pethema after a median follow-up
of 66 months [abstract]. Blood. 2003;102:42a) |
164 |
< 65 |
11 vs 30 |
0.002 |
34 vs 42 |
NS |
67 vs 65 |
NS |
US Inter Group (USIG) (Barlogie B, Kyle R, Anderson K, et al. Comparable
survival in multiple myeloma with high dose therapy employing Mel 140mg/m2
+ TBI 12 Gy autotransplants versus standard dose therapy with VBMCP and
no benefit from interferon maintenance: results of Intergroup trial S9321
[abstract]. Blood. 2003;102:42a) |
50 |
|
15 vs 17 |
NS |
21 vs 25 |
0.05 |
53 vs 58 |
NS |
Italian Multiple Myeloma Study Group (IMMSG) (Palumbo A, Bringhen S,
Petrucci MT, et al. A prospective randomized trial of intermediate dose
melphalan (100 mg/m2) vs oral melphalan/prednisone [abstract]. Blood. 2003;102:984a) |
195 |
50-70 |
7 vs 26 |
< 0.0001 |
16 vs 28 |
0.0036 |
43 vs 58+ |
0.0008 |
CR defined either by a negative immunofixation (true CR) or by a negative
electrophoresis (near CR) was significantly more frequent after HDT, except
in the US Intergroup Study where the CR rate obtained with CC was higher
than expected (15%). Median EFS was always longer with HDT compared to
CC (4-12 months). The difference was significant in 5/6 trials (except
in the Spanish trial, where patients refractory to initial CC were excluded).
The median EFS is remarkably constant (25-31 months). It is more difficult
to analyze OS results since OS partly depends on salvage therapy. However,
median OS was significantly longer in the three studies where differences
in EFS were more marked. In all studies procedure-related death rate was
< 5% and not greater than that observed with CC. From this large experience
one can now conclude that HDT supported by ASCT is the standard of care
of patients with newly diagnosed MM, at least up to the age of 65. High-dose
methotrexate
(HDM) 200 mg/m2 should be preferred to HDM 140 mg/m2
plus TBI 8-12 Gy delivered in 4 fractions as the conditioning regimen for
ASCT in MM; total marrow irradiation (9 Gy), busulfan
(12 mg/kg) and cyclophosphamide
(120
mg/kg) is another possibilityref
-
consolidation chemotherapy (CC) after single autologous peripheral
blood stem cell transplant (auto-PBSCT) in 103 mostly newly diagnosed myeloma
patients (67 patients were < or =6 months from the initial treatment).
Patients received conditioning with BCNU, melphalan+/-gemcitabine and auto-PBSCT
followed by 2 cycles of the DCEP+/-G regimen (dexamethasone, cyclophosphamide,
etoposide, cisplatin+/-gemcitabine) at 3 and 9 months post-transplant and
alternating with two cycles of DPP regimen (dexamethasone, cisplatin, paclitaxel)
at 6 and 12 months post-transplant. With a median follow-up of 61.2 months,
the median event-free survival (EFS) and overall survival (OS) are 26 and
54.1 months, respectively. The 5-year EFS and OS are 23.1 and 42.5%, respectively.
Overall, 51 (49.5%) patients finished all CC, suggesting that a major limitation
of this approach is an inability to deliver all planned treatments. In
order to improve results following autotransplantation, novel agents or
immunologic approaches should be studied in the post-transplant settingref
-
tandem autologous transplantation : in the IFM90 trial, achievement
of a least a VGPR (> 90% reduction of the M-component) was significantly
correlated with longer survival. The impact of achieving CR on the duration
of disease control and survival was confirmed by several investigatorsref.
Thus, in MM as in other hematological malignancies, the primary objective
of therapy should be to achieve CR. One way to increase the CR rate is
to repeat intensive treatments. To this end, Barlogie and colleagues in
Arkansas used a strategy with tandem ASCTref
that appeared to increase the CR rate to approximately 40%. In newly diagnosed
patients this better tumor cell reduction was translated into encouraging
EFS and OS of 43 months and 68 months, respectivelyref.
However, comparison with less aggressive strategies is needed to evaluate
the actual impact of tandem ASCT on EFS and OS. The IFM was again the first
to conduct a randomized trial (IFM94) comparing single and double ASCTref.
In this study, 399 previously untreated patients under the age of 60 years
were randomly assigned to receive either a single ASCT prepared by melphalan
140 mg/m2 (Mel 140) plus total body irradiation (TBI) or
a double ASCT, the first being prepared by Mel 140 and the second by Mel
140 plus TBI. On an intention-to- treat basis, CR or very good partial
remission was achieved by 42% of patients in the single ASCT group versus
50% in the double ASCT group (P = 0.10). The probability of 7-year EFS
was 10% versus 20% (P = 0.03) and the probability of 7-year OS was
21% versus 42%, respectively. These results are confirmed by a preliminary
analysis of a comparable study conducted by the Bologna Group (Cavo M,
Zamagni E, Cellini C, et al. Single versus tandem autologous transplants
in multiple myeloma: Italian experience (abstract). The Hematol J. 2003;4:supp
1:560). The preliminary analysis of a Dutch trial reported by Segeren and
colleagues yielded negative results, in that there was no improvement in
outcome with more intensive therapyref.
However, the updated results showed a significant advantage in term of
EFS in the more intensive treatment group (Sonneveld P, Van Der Holt B,
Segeren CM, et al. Intensive versus double intensive therapy in untreated
multiple myeloma. Updated analysis of the prospective phase III study Hovon
24-MM [abstract]. The Hematol J. 2003;4, suppl 1:559-560). 2 other studies
have been presented in part in meetings (Fermand JP, Alberti C, Marolleau
JP. Single versus double high dose therapy supported with autologous blood
stem cell transplantation using unselected or CD34 enriched ABSC: results
of a two by two designed randomized trial in 230 young patients with multiple
myeloma [abstract]. The Hematol J. 2003;4, suppl 1:S59; Goldschmidt H.
Single versus double high dose therapy in multiple myeloma: second analysis
of the trial GMMG-HD2 [abstract]. Proc Multiple Myeloma 2004 Meeting. Torino,
Italy; 22-24 April 2004: 119). Although the CR rate is significantly higher
in only 1 trial, the median EFS is significantly longer in 4/5 trials.
A significant difference in OS was found only in the IFM94 trial but, since
divergence of the OS curves occurred only after 4 years, longer follow-up
is probably needed before drawing definite conclusions from other trials.
The available data are in favor of tandem autotransplants. Results of randomized
studies of single versus double autologous HSCT :
study
|
no. of pts
|
preparative regimen
|
CR rate (%)
|
EFS (months)
|
OS (months)
|
age
|
single
|
double
|
difference
|
single vs
double
|
p
value
|
single vs
double
|
p
value
|
single vs
double
|
p
value
|
Intergroupe Francophone du Myelome (IFM94)ref |
339 |
< 61 |
Mel140+TBI |
Mel280+TBI |
Mel140 |
42 vs 50 |
0.10 |
25 vs 30 |
0.03 |
48 vs 58 |
0.01 |
Myelome Auto Greffe (MAG) 95 (Fermand JP, Alberti C, Marolleau JP.
Single versus double high dose therapy supported with autologous blood
stem cell transplantation using unselected or CD34 enriched ABSC: results
of a two by two designed randomized trial in 230 young patients with multiple
myeloma [abstract]. The Hematol J. 2003;4, suppl 1:S59) |
227 |
< 56 |
Mel140+
multidrug
+ TBI |
Mel280+
VP16+TBI |
< Mel140 |
39 vs 37 |
NS |
31 vs 33 |
NS |
49 vs 73 |
0.14 |
Bologna (Cavo M, Zamagni E, Cellini C, et al. Single versus tandem
autologous transplants in multiple myeloma: Italian experience (abstract).
The Hematol J. 2003;4:supp 1:560) |
220 |
< 61 |
Mel200 |
Mel320+
BU12 |
Mel120+
BU12 |
31 vs 43 |
NS |
21 vs 31 |
0.02 |
56 vs 60 |
NS |
German Multiple Myeloma Group (GMMG) (Goldschmidt H. Single versus
double high dose therapy in multiple myeloma: second analysis of the trial
GMMG-HD2 [abstract]. Proc Multiple Myeloma 2004 Meeting. Torino, Italy;
22-24 April 2004: 119) |
261 |
< 66 |
Mel200 |
Mel400 |
Mel200 |
- |
- |
23 vs NR |
0.03 |
- |
- |
Hovon (Sonneveld P, Van Der Holt B, Segeren CM, et al. Intensive versus
double intensive therapy in untreated multiple myeloma. Updated analysis
of the prospective phase III study Hovon 24-MM [abstract]. The Hematol
J. 2003;4, suppl 1:559-560) |
303 |
< 66 |
Mel140 |
Mel140+
Cy120+TBI |
Cy120+TBI |
13 vs 28 |
0.002 |
20 vs 22 |
0.01 |
55 vs 50 |
NS |
As compared with a single infusion of stem cells, 2 infusions significantly
prolonged both OS and EFS, particularly among patients who did not enter
remission after receiving the first transplantref1,
ref2.
The benefit of melphalan-based double autologous transplantations was greatest
in patients whose myeloma cells had a normal karyotype: these patients
had an estimated probability of remaining in remission at 10 years of nearly
20%, suggesting that long-term control and even cure of MM are possible
with double autologous stem-cell transplantationref.
recent large trials of double autologous transplants yielded feasibility
rates of 70 to 80%ref1,
ref2
After 7 years, 42% of patients who got
2 rounds of HDT followed each time by a HSCT were still alive, compared
with 21% of those who received the standard single round of chemo plus
a transplant. In addition to increasing life span, the second round of
treatment doubled the chance of surviving 7 years without a recurrence
of cancer (20% versus 10%). The difference was most striking for people
who had not had a good response to the first transplant. Only 11% of those
patients lived 7 years without a second transplant. With a second transplant,
43% survived that long. The combination of high levels of b2m
and chromosome 13 deletion allows identification of a high-risk subgroup
of patients with de novo MM. In this population of patients, we
have evaluated the impact of a murine anti-IL-6
mAb (BE-8)
as part of the second conditioning regimen in a multicenter prospective
randomized trial of tandem autologous stem cell transplantation (ASCT).
Conditioning for the first ASCT was accomplished with melphalan 200 mg/m2
and for the second one with melphalan 220 mg/m2 + dexamethasone
with or without BE-8 infusion. This trial included 219 patients, of whom
166 were randomized, 85 without BE-8 (arm A) and 81 with BE-8 (arm B).
The median OS and EFS times of the whole group of patients were 41 and
30 months, respectively. Response rates, OS, and EFS were not different
between the 2 arms of the trial. OS at 54 months was 46% in arm A and 51%
in arm B (P = .90); median EFS was 35 months in arm A and 31 in
arm B (P = .39). In high-risk patients the dose intensity of melphalan
at 420 mg/m2 led to encouraging results, but the addition of
anti-IL-6 mAb to the second conditioning regimen did not improve either
OS nor EFSref.
In 53 South Korean patients with MM who received autologous HSCT from
April 1996 to September 2004, a second HSCT is the most significant factor
for an improved PFS and OS after the first ASCT (P < 0.001, respectively).
Up-front double SCT is needed to improve the OS and PFS in patients with
MMref.
Between August 1993 and March 2003, 130 consecutive MM patients eligible
for high-dose treatment were offered a program including up-front autologous
HSCT after conditioning with 200 mg/m2 melphalan followed by
a second ASCT in case of relapse or progression. A total of 107 (82%) patients
completed the first ASCT. The best response obtained after ASCT was CR
23%, VGPR 28%, PR 42%, and minimal response (MR) 7%. Median OS and EFS
were 65.4 and 27.7 months, respectively. Relapse or progression occurred
in 70 patients; 26 received a second autologous HSCT (with a median time
of 20.4 months from first ASCT). A major response (> or =PR) was obtained
in 69% of these patients. Median OS and EFS after the second ASCT were
38.1 and 14.8 months. TRM was 1.9% after the first autologous HSCT but
no deaths occurred after the second. Elective up-front single ASCT followed
by second ASCT after relapse or progression is a safe and effective
global strategy to treat MM patientsref.
Tandem autologous transplants with suboptimal dosing are inferior even
to oral melphalan-prednisone (MP) + thalidomide (THAL) (Facon T, Mary JY,
Hulin C, et al. Major superiority of melphalan-prednisone (MP) + thalidomide
(THAL) over MP and autologous stem cell transplantation in the treatment
of newly diagnosed elderly patients with multiple myeloma. Blood 2005;106:230a-230a)
-
prognostic factors in the context of autologous HSCT
: as with CC, a number of parameters have been shown to significantly influence
the outcome after ASCT in newly diagnosed patientsref1,
ref2.
-
standard biological markers that are important predictors of outcome after
CC has also been demonstrated for ASCT.
-
high levels of ß2Mref
-
high levels of C-reactive protein
-
high levels of LDH
-
low levels of albumin
-
age
-
response to therapyref
-
high bone marrow plasma-cell labeling index (PCLI)
-
genetic abnormalities as evaluated by conventional cytogenetics or by molecular
biology techniques appear to be powerful prognostic markers as well. Based
on a large experience with tandem ASCT, statistical analysis performed
by the Arkansas group has shown that hypodiploidy and cytogenetic abnormalities
such as chromosome 13 deletionsref1,
ref2
or myelodysplastic-like abnormalitiesref
are associated with a poor prognosisref1,
ref2
-
del13qref
either by conventional cytogeneticsref1,
ref2
or by FISHref
is an adverse prognostic factor. While metaphase-defined chromosome 13q
deletion is found in only 15% of karyotypes, FISH detects this abnormality
in approximately 45% of patients. However, when the deletion is found by
conventional cytogenetics, the prognosis appears to be even poorerref.
-
translocations involving 14q32 has also been studied retrospectively
-
t(4;14)(p16.3;q32) : by FISH analysis, is present in approximately
13% of patients and is associated with a short EFS and OSref1,
ref2.
-
t(4;14)(p16.3; q32) identified by elevated cyclin D1 levels as evaluated
by real-time RT-PCR appear to enjoy longer remissions and to have longer
survivalref1,
ref2
-
-17p13
-
presence of circulating myeloma cells (CMCs) does not predict for
chemosensitivity but rather for short time to progression perhaps in part
due to a higher proliferation rate as reflected by the higher PCLI. The
poor outcome with CMCs should not be interpreted as justification for purging
of the apheresis product since there is no evidence that purging of the
HSC prior to transplantation confers any benefitref.
CMCs may not be involved in posttransplantation relapse but may reflect
loss of adhesion indicative of unfavorable biology. CMCs can be detected
by :
-
slide-based immunofluorescence method has been shown to predict early relapse
of the disease after HDT/ASCTref1,
ref2.
However, this test is time-consuming, technically difficult, and not widely
available.
-
flow cytometry is widely available and can detect CMCsref.
Detection of circulating myeloma cells at the time of diagnosis of multiple
myeloma is associated with a shortened survivalref1,
ref2.
The presence of CMCs is not a consequence of disease burden but suggests
biologically aggressive diseaseref.
Thus, detection of CMCs by flow cytometry prior to HSC collection would
be a predictor of biologically aggressive disease leading to early relapse
regardless of initial response to HDT/ASCT therapy.
There are no differences in tumor load and hematopoietic stem cell yield
in first and second apheresis, offering the possibility of combining LP
harvested over several days without increasing the tumor load per CD34+
cellref.
Relapse risk after autologous HSCT in patients with newly diagnosed MM
is not related with infused tumor cell load and the outcome is not improved
by CD34+ cell selection: long term follow-up of an EBMT phase
III randomized studyref
-
achieving a CR is considered an important goal of HDT/ASCT, since
various studies have shown that achieving a CR is associated with an improved
TTP and OSref1,
ref2,
ref3,
ref4.
However, achieving a CR is not the only factor associated with an improved
survival, since CR rates with single and tandem transplantations were not
significantly different in the IFM 94 trial, yet patients who underwent
tandem transplantation had an improved survivalref.
The lack of correlation between CR and OS and TTP has been reported in
a smaller cohort of patientsref
For instance, Barlogie and colleagues in Arkansas have shown that patients
with low ß2M (or low LDH) and no cytogenetic abnormalities
have a good prognosis when treated with tandem ASCTref1,
ref2.
Patients with high ß2M and cytogenetic abnormalities (especially
hypodiploidy and/or chromosome 13 deletion) have a poor prognosis even
with tandem ASCT, and for these patients new treatments are clearly neededref1,
ref2.
The IFM group has shown similar prognostic implications for the combination
of ß2M and chromosome 13 deletions detected by FISH analysis.
Patients with low ß2M and no chromosome 13 abnormality
by FISH had a median OS of 111 months, while patients with high ß2M
levels and chromosome 13 deletions had a median OS of only 25 months. These
findings led to the risk-adapted IFM99, with 2 different strategies for
standard-risk patients (0 or 1 adverse prognostic factor) and for high-risk
patients (2 adverse prognostic factors). The use of initial prognostic
factors could also help in determining which patients benefit more from
HDT. Unfortunately, in most randomized trials, the number of patients is
too small to evaluate the impact of HDT in each prognostic subgroup. In
the MRC7 trial comparing CC and HDT, there was a trend toward a greater
survival benefit with HDT for the group of patients with a poor prognosis
defined by a high ß2M level (> 8 mg/L)ref.
In the IFM94 trial, double ASCT was superior to single ASCT in prognostic
subgroups defined by ß2M or LDH but, due to a small number
of patients in each group, the difference was not statistically significant.
The superiority of double ASCT was only significant in patients failing
to achieve at least 90% reduction of the M-component within 3 months after
one ASCTref.
Therefore, according to available data HDT could be proposed to all patients
and could be even more beneficial for patients with adverse prognostic
factors. Blade and colleagues reported that there was no significant difference
between CC and HDT in patients responding to initial CC in the Spanish
Cooperative Group PETHEMA randomized trial (Blade J, Sureda A, Ribera JM,
et al. High-dose therapy/autotransplantation/intensification versus continued
conventional chemotherapy in multiple myeloma patients responding to initial
chemotherapy. Definitive results from Pethema after a median follow-up
of 66 months [abstract]. Blood. 2003;102:42a), suggesting that patients
who fail to respond to their first treatment should undergo ASCT as well.
211 MM patients received autologous PBSCT at Hammersmith Hospital between
1994 and 2004 after conditioning with melphalan alone (183 patients), or
melphalan and total blood irradiation (28 patients). The influence of age,
type of MM, status prior and post ASCT, previous treatment regimens, time
of ASCT from diagnosis, year of autograft, dose of re-infused CD34+
cells, plasma cell infiltration and b2-microglobulin
at diagnosis on overall survival (OS) and event-free survival (EFS) were
evaluated to define patients with better prognosis. Median OS and EFS from
transplantation were 50.9 and 20.1 months, respectively. Median OS from
diagnosis was 68.8 months. TRM was 1.4%. Lower b2-microglobulin
levels, achievement of CR post transplant and lower plasma cell infiltration
at diagnosis and transplant correlated with longer EFS and OS, whereas
CR at transplant and low IPI at transplant correlated with better EFS.
Higher CD34+ cell dose correlated with improved OSref.
-
how to further improve the outcome of ASCT
: in the IFM94 trial, the 7-year EFS following ASCT is only 20%
-
improve pretransplant chemotherapy regimens :
-
induction :
-
effect of chemotherapy with alkylating agents on the yield of CD34+
cells : 789 patients included in the Spanish multicenter protocol GEM-2000
underwent mobilization therapy after 4 courses of alternating VBMCP/VBAD
chemotherapy. The mobilization regimens consisted of standard or high-dose
G-CSF in 551 (70%) patients, and chemotherapy and G-CSF in 206 (26%) patients.
The CD34+ cell yield was < 4x106/kg in 388 patients
(49%), and equal or greater than 4x106/kg in 401 patients (51%).
Multivariate analysis indicated that advanced age (p<0.0001) and longer
interval between diagnosis and mobilization (p=0.012) were the two variables
associated with a lower CD34+ cell yield. Significant differences
in CD34+ cell yield were not observed between the mobilization
regimens. Of the 789 patients included in the protocol, 726 (92%) underwent
the planned ASCT, whereas 25 (3%) patients did not because of the low number
of CD34+ cells collected. Following ASCT, 0.5x109
neutrophils/L could be recovered after 11 days (median time; range, 5-71
days) and 20x109 platelets/L could be recovered after 12 days
(median time; range, 6-69 days). A short-course of therapy with alkylating
agents according to the GEM-2000 protocol was associated with an appropriate
CD34+ cell collection, and allowed the planned ASCT to be performed
in the majority of MM patientsref
-
improve conditioning :
-
melphalan
200 mg/m2 (Mel 200) is considered as the standard preparation
regimen prior to ASCTref.
In the absence of randomized trials, the benefit of other regimens using
higher doses of melphalan, combinations of drugs, or melphalan coupled
with bone-seeking radioisotopes remains unknown. It is clear, however,
that in the context of tandem ASCT, the dose intensity of the preparative
regimen is crucial. In the IFM94 trialref
the CR rate was not significantly higher in the double ASCT arm, but the
preparative regimen prior to the first ASCT was only Mel 140, while in
the Arkansas experience the CR rate appears higher with 2 ASCT prepared
by Mel 200 (Barlogie B, Kyle R, Anderson K, et al. Comparable survival
in multiple myeloma with high dose therapy employing Mel 140 mg/m2
+ TBI 12 Gy autotransplants versus standard dose therapy with VBMCP and
no benefit from interferon maintenance: results of Intergroup trial S9321
[abstract]. Blood. 2003;102:42a).
-
melphalan-based autologous HSCT is a standard therapy for multiple myeloma,
but is associated with severe oral mucositis (OM). To identify predictors
for severe OM, we studied 381 consecutive newly diagnosed myeloma patients
who received Mel-ASCT. Melphalan was given at 200 mg/m2 body
surface area (BSA), reduced to 140 mg/m2 for serum creatinine
>3 mg/dl. Potential covariates included demographics, pre-transplant serum
albumin and renal and liver function tests, and mg/kg melphalan dose received.
The BSA dosing resulted in a wide range of melphalan doses given (2.4-6.2
mg/kg). OM developed in 75% of patients and was severe in 21%. Predictors
of severe OM in multiple logistic regression analyses were high serum creatinine
(odds ratio (OR)=1.581; 95% confidence interval (CI): 1.080-2.313; P=0.018)
and high mg/kg melphalan (OR=1.595; 95% CI: 1.065-2.389; P=0.023). An OM
prediction model was developed based on these variables. BSA dosing of
melphalan results in wide variations in the mg/kg dose, and that patients
with renal dysfunction who are scheduled to receive a high mg/kg melphalan
dose have the greatest risk for severe OM following Mel-ASCTref
-
"Total Therapy I"
-
"Total Therapy II" (Barlogie B, Jacobson J, Sawyer J, et al. Increasing
CR frequency as a strategy toward extending event-free survival and overall
survival in multiple myeloma: 4-year results of Total Therapy II versus
Total Therapy I (abstract). Blood. 2003;102:42a). This is an aggressive
protocol with 4 consecutive phases:
This trial also addressed the possible contribution of thalidomide
in a randomized trial design from initiation of treatment, but the thalidomide
arm is still blinded. Current analysis of 462 patients included into Total
Therapy II program compared to 231 patients treated with the previous "Total
Therapy I" protocol shows a higher CR rate (66% versus 43%) and an increased
4-year EFS rate. This improvement was more marked for patients without
cytogenetic abnormalities (4-year EFS 70% versus 37%). The 5-year OS
rate was 68% among 532 patients under 65 years of ageref,
which is virtually identical to the outcome in the autograft–allograft
group reported by Bruno and colleagues without the unpredictable consequences
of an incidence of at least 30% of extensive cGvHDref
-
"Total Therapy 3" : incorporation of bortezomib
into tandem-autograft program has improved survival in high-risk subgroupsref
-
high-dose idarubicin
42 mg/m2 + melphalan
200 mg/m2 + cyclophosphamide
120 mg/kg as conditioning for autologous HSCT increases TRM compared
to melphalan
200 mg/m2 in patients with MMref
-
153Sm-EDTMP
in myeloablative dosage followed by a second autotransplantation in patients
with relapsed MMref
-
2 courses of pulsed-VAD
(vincristine, adriamycin, dexamethasone)
followed by 2 courses of DCEP
(dexamethasone, cyclophosphamide, etoposide and cis-platinum)
has an adequate mobilizing capacity, without significant toxicity, and
a good anti-myeloma activity (a higher percentage of responses (CR+VGPR+PR)
than VAD alone), and therefore represents a safe and effective therapeutic
approach for multiple myeloma patients at the onset of their diseaseref
-
high-dose cyclophosphamide
is often used before ASCT for mobilization of HSCs. The Mayo Clinic MM
transplantation database was searched for patients who had HSC mobilization
with G-CSF alone or cyclophosphamide + G-CSF. The impact of cyclophosphamide
on CR rates and TTP was evaluated. A cohort of 201 patients was identified:
127 mobilized with cyclophosphamide + G-CSF and 74 with G-CSF alone. There
were no statistically significant differences between the 2 cohorts in
regard to age, sex, b2-microglobulin
level, plasma cell labeling index, cytogenetics, conditioning regimen,
or disease status at time of HSCT. CR rates were 37.4% and 41.3% (P = 0.6115)
for patients mobilized with cyclophosphamide + G-CSF and G-CSF alone, respectively,
and TTPs were 19.9 months and 20.9 months (P = 0.59). In a multivariate
analysis for TTP, cytogenetics and CR rates were the only independent variables
(P = 0.0012 and P < 0.0001, respectively)ref
-
busulfan
followed by melphalan
: 44 consecutive patients with MM participating in the co-operative Spanish
protocol were prospectively evaluated. BU pharmacokinetic parameters were
estimated for each patient after the first dose based on measured concentrations
and subsequent doses were modified as necessary to achieve target exposure.
Mean BU exposure (AUCss) (+/-DS) before dosage modification
range from 3192 to 12 180 ng h/mL. 26 out of 44 (59%) patients required
dose adjustment. None of the patients developed hepatic veno-occlusive
disease (VOD). Grade > or = II oropharyngeal mucositis was observed in
the majority of patients (95%) and the severity of mucositis increased
with increasing average steady-state BU plasma concentration. There were
4 treatment-related deaths: 2 patients died from multiorgan failure and
two of respiratory infections. Of the remaining 40 patients, 15 were in
CR with negative immunofixation, 21 in partial remission and four in stable
disease 3 months after ASCT. The results of the present study show the
variability in BU pharmacokinetic parameters and suggest the possible relationship
between toxicities and BU exposure. Individualising BU dosage in MM patients
undergoing ASCT we observed the absence of VODref
-
intermediate-dose melphalan
(100 mg/m2)/bortezomib
/thalidomide
/dexamethasone
in patients with refractory or relapsed MM : bortezomib and thalidomide
have shown synergy with melphalan and dexamethasone. 26 patients with advanced-stage
myeloma were treated with melphalan 50 mg/m2 and bortezomib
1.3 mg/m2 on days -6 and -3 in association with thalidomide
200 mg and dexamethasone 20 mg on days -6 through -3, followed by HSCT
on day 0. Nonhematologic toxicities included pneumonia, febrile neutropenia,
and peripheral neuropathy. All patients had undergone autologous HSCT at
diagnosis, and 13 patients (50%) underwent an additional transplantation
at relapse. Responses occurred in 17 of 26 patients (65%), including 1
complete remission, 3 near complete remissions (12%), and 2 very good partial
remissions (8%). Response rate was higher than that induced by the previous
line of treatment in 12 patients (46%)ref
-
skeletal
targeted radiotherapy (STR)

-
166Ho-DOTMP
: 83 patients received high dose 166Ho-DOTMP followed by melphalan
and transplant of PBSCs. 25 patients also received 8 Gy total body radiation
(TBI). Dosages administered ranged from 460 to 4476 mCi 166Ho-DOTMP.
Marrow dose was derived using the assumption that all radioactivity not
excreted by 20 hours was localized to the bone surfaces, and applying the
Eckerman bone and marrow dose model to the calculated bone residence times.
The dosimetry of the urinary bladder and kidneys was important because
of the rapid excretion of the non-targeted radioactivity via the urinary
pathway. The dynamic bladder model was used for bladder wall surface dose,
and the ICRP 53 kinetic model was used to model kidney kinetics with an
additional blood component included. Marrow doses ranged from 13 to 59
Gy and successful hematapoietic recovery occurred. Bladder doses ranged
from 4.7 to 157 Gy. Hemorrhagic cystitis occurred in some patients who
received more than 40 Gy to the bladder wall surface. Bladder irrigation
was successful in protecting patients from bladder toxicity. Kidney doses
ranged from 0.5-7.9 Gy. Kidney toxicity in the form of thrombotic microangiopathy
with renal dysfunction was observed, with the severity being related to
Ho-166-DOTMP radiation dose and probably the dose rate as well. In a future
trial, kidney dosimetry will be assessed using early serial g-camera
imaging and modifications will be implemented to reduce renal toxicityref1,
ref2,
ref3,
ref4
-
153Sm-EDTMP
:12 patients were treated with escalating doses of 153Sm-EDTMP (N=3/group;
6, 12, 19.8, and 30 mCi/kg) and a fixed dose of melphalan (200 mg/m2).
No dose limiting toxicity was seen. To better standardize the marrow compartment
radiation dose, the study was modified such that an additional six patients
were treated at a targeted absorbed radiation dose to the red marrow of
40 Gy based on a trace labeled infusion 1 week prior to the therapy. Despite
rapid elimination of unbound radiopharmaceutical via kidneys and bladder,
no episodes of nephrotoxicity, hemorrhagic cystitis, or delayed radiation
nephritis were observed with a median follow-up of 31 months (range 8.5-44).
Median times to ANC>0.5 and platelet >20 x 106/l were 12 and
11 days, respectively, with no graft failures. ORR was 94% including 7
VGPRs and 5 complete responses. Addition of 153Sm EDTMP to melphalan conditioning
appears to be safe, well-tolerated and worthy of further studyref
-
allogeneic HSCT
ref1,
ref2
appear to offer a better chance for a possible cure of myeloma thanks to
GVT
effect
(here renamed graft-versus-myeloma (GVM) effect) : if given early
in the course of the disease, allogeneic BMT yields molecular remissions
and about 33% of the patients remain free of disease 6 years later) but
...
-
it is available only for a small minority of MM patients under 50 years
of age and having a HLA identical sibling
-
it has been associated with an unusually high transplantation-related mortality
(TRM) mainly as a consequence of
-
bacterial and fungal infections => advent of less toxic non-myeloablative
BMT (miniallo-transplant)
(conditioning regimen =
-
low-dose TBI
-
melphalan
:
-
MEL100 (100 mg/m2) : CR = 35%, EFS = 32 months; significantly
reduced haematological and extra-haematological toxicity; tandem MEL100
was less toxic than tandem MEL200
-
MEL200 : CR = 48%; EFS = 42 months; OS and TRM were not different
... combined with donor
leukocyte infusion (DLI)
has provided an 81% short-term survival in a trial combining this approach
with an initial conventional autologous
BMT
to reduce tumor burden (as results are related to diseases status and relapse
rate is high in patients with advanced disease). Level of chimerism did
not correlate with either the quality of response or aGVHD. No significant
differences were found between bone marrow and peripheral blood samples.
Analogously, even if donor DNA percentage often resulted higher in the
PMN fraction than in the mononuclear one, these differences were not significant
after statistical analysis. On the other hand, cGVHD was associated with
increased rates of FDCref
-
GvHD => advent of CD6
T-cell
depleted (TCD) allogeneic BMT reduces the incidence of GvHD but is
associated to a higher relapse rate due to abrogation of GVT
effect
,
which can ayway be maintained with prophylactic CD4+ donor
leukocyte infusion (DLI)
administered 6 to 9 months after BMTref.
Between 1989 and 2000, 71 patients received a myeloablative unrelated transplant
for multiple myeloma; 70 patients consented for this analysis. The median
recipient age was 44 years. A total of 31% of patients had received a prior
autologous transplant. In all, 91% of patients engrafted. The 3-year cumulative
incidence estimate of relapse was 34+/-10%. The incidence of grade II-IV
GVHD was 47%. The Kaplan-Meier estimate for overall survival at 5 years
was 9+/-7%. The 100-day treatment-related mortality was 42%ref.
-
high relapse rates
The use of allogeneic
peripheral blood precursor cell (PBPC) trasplant (PBSCT)
instead of BMT is associated with earlier engraftment but yields no significant
survival benefits.
-
standard (myeloablative) conditioning regimen still has a limited
role in MM. Because of its toxicity, it can be proposed only to a small
number of patients with MM (patients aged < 55 years with an HLA-identical
sibling). Even in younger patients TRM is very high with standard regimens,
mainly as a consequence of infection and GVHD. However, there are several
arguments in favor of pursuing allogeneic SCT in MM:
-
analysis of the EBMT Registry showed improved results in the late 1990s,
mostly due to a higher frequency of early transplantationref
-
comparison of autologous and allogeneic transplantations show that short-term
survival is superior with autologous SCT, mainly due to a higher transplant-related
mortality in the first year with allogeneic SCT. However, while patients
treated with autologous SCT can experience late relapses, there is a plateau
of the EFS and OS curves after 5 years for patients receiving allogeneic
transplantation. Long-term survival could be superior with allogeneic HSCTref.
-
the so-called GvM effect has been demonstrated by the efficacy of donor
lymphocyte infusions in patients relapsing after allogeneic BMTref.
Therefore, recent clinical studies on allogeneic HSCT in MM have focused
on frontline therapy with the goal of reducing TRM while harnessing the
GvM effect. One way to influence TRM is to reduce the incidence and severity
of GVHD by TCD. While attempts to reduce GVHD have frequently been associated
with a higher incidence of relapse, preliminary results of prophylactic
CD4+ DLIs after CD6 T cell depletion appeared promisingref.
But it has been recently shown that response to DLIs is statistically related
to the occurence of GVHD, suggesting that in MM, like in other hematological
malignancies, targets for graft versus host and graft versus myeloma are
identical or that antitumor activity is related to GVHDref.
Finally, results of a Dutch prospective evaluation of T cell-depleted allogeneic
SCT as part of frontline therapy were very disapointingref.
In this series of 53 patients up to the age of 55 with an HLA-identical
sibling, the CR rate was only 19% and median OS from transplantation was
only 25 months. The cause of death was either disease progression or transplant-related
toxicity.
In the Intergroup Trial S9321ref,
the study design included myeloablative allografting for patients 55 years
of age or younger with a suitable sibling donor. The treatment option was
discontinued early in this group of patients because of a TRM rate of 53%.
However, 22% of the patients in this group were alive and free of disease
progression 7 years after allografting. Garban et al. did not find an advantage
of allografting over melphalan-based autografting in patients with high-risk
myeloma (i.e., those with an elevated b2-microglobulin
level plus chromosome 13 abnormalities)ref.
That study did not enroll patients who were at intermediate or good risk,
nor were additional prognostic factors evaluated by intention-to-treat
analysis. Furthermore, the potent pretransplantation immune suppression
with ATG used in the study of Garban et al. may have prevented GvM effects.
Neither chromosome 13 abnormalities nor b2-microglobulin
levels appeared to affect the outcome after allografting.
Plasma cell chimerism after allogeneic HSCT is delayed in comparison
to T-cell chimerism : increasing and stable chimerism were associated
with ongoing remission in 15 out of 16 samples (93%), and decreases in
chimerism predicted relapse in 5 out of 6 patientsref
-
hydroxyurea, cytarabine, busulfan, cyclophosphamide, and semustine.
10 patients underwent allogeneic transplantation among them hydroxyurea
(40 mg/kg) was administered twice on day-10 and cytarabine (2 g/m2)
was given on day-9, busulfan was administered orally in four divided doses
daily for 3 days (days-8 to -6). The dose of busulfan was 12 mg/kg in the
protocol followed by cyclophosphamide intravenously over 1 hour on days-5
and -4 (1.8 g/m2), and with semustine (Me-CCNU) 250 mg/m2
on day -3. Chimerism data were available on all patients and all patients
achieved full donor chimerism without graft failure. 6 patients had not
acute graft-versus-host disease (GVHD, 36.4%; 95% CI: 13.9% - 38.6%). 2
patients (18.2%) developed grade I acute GVHD (95% CI: 10.9% - 35.9%) and
grade II acute GVHD occurred in one patient (9.1%; 95% CI: 8.4% - 32.3%).
Severe grade IVa GVHD was seen in one patient, who died from acute GVHD.
The incidence of chronic GVHD was 22.2% (95% CI: 11.7% - 36.7%), among
them one died of severe grade IV GVHD and one developed multiorgan failure
on day +170; the treatment-related mortality was 22.0% (95% CI: 10.3% -
34.1%). The overall 4-year survival rate was 67.8% (95% CI: 16.3% - 46.7%).
The estimated 4-year progression-free survival rate was 58.5% (95% CI:
13.7% - 41.8%). The 4-year complete remission was 72.7% (95% CI: 27.8%
- 49.6%). One patient relapsed after 4 months and achived the complete
remission after receiving the DLIref.
-
reduced-intensity allogeneic transplantation : nonmyeloablative
or reduced-intensity allogeneic SCT (so-called miniallogeneic transplantation)
represents a new hope and a new way of taking advantage of the graft-versus-myeloma
effect related to the donor immune system while reducing transplant-related
toxicity. However, it has been shown that relapses are frequent when reduced-intensity
regimens are used in patients with relapsed/refractory diseaseref1,
ref2.
In the past few years, reduced-intensity regimens have been widely used
after tumor burden reduction with HDT + ASCT. The feasibility of reduced-intensity
allogeneic transplantation after one or two ASCT has been shownref.
HDT with melphalan 200 mg/m2 supported by ASCT yields a high
response rate with a low toxicity and a low transplant-related mortality.
Allogeneic SCT with nonmyeloablative conditioning regimens provides a tumor-free
stem cell source and reduces transplant-related mortality while harnessing
the graft-versus-myeloma effect. Although the risk of acute GVHD and of
infections related to chronic GVHD and its treatment remains, this approach
appears to be safer than allogeneic BMT and can be proposed to older patients
(up to 65 years of age). 2 groups have published their preliminary experienceref1,
ref2
:
author
|
patients
|
age
(median)
|
autologous
SCT
|
RIC
|
GVHD
|
TRM
|
CR rate
|
median follow-up
|
survival
|
Maloneyref |
54 (48% relapsed or refractory MM) |
29-71 (52) |
Mel 200 |
low-dose*
TBI (2 Gy ) |
38.5% aGVHD II
46% cGVHD |
100 days 2%
1-yr 15% |
52% |
18 m |
2-yr EFS 55%
2-yr OS 78% |
Krogerref |
47 (23 unrelated donors) |
31-64 (52) |
Mel 200 |
Flu/Mel/ATG |
32% aGVHD II
32% cGVHD |
100 days 6% |
55% |
15 m |
3-yr EFS 54%
3-yr OS 70% |
Gerullref |
52 patients with relapsed MM or high-risk features at diagnosis received
. Regimen-related toxicity was low. . |
patients were heavily pretreated with a median of 8 cycles of conventional
chemotherapy and one or more autologous transplants for all but one patient |
|
2 Gy TBI alone (n=3) or with fludarabine (n=49) as conditioning |
aGvHD II-IV occurred in 37% of patients, and 70% experienced cGvHD |
567 days 17% |
|
|
Estimated PFS and OS at 18 months was 29.4 and 41.1%, respectively.
Patients with cGvHD had a significantly higher PFS, as did patients with
up to 8 cycles of pretreatment chemotherapy vs those with >= 9. In this
highly pretreated patient group, disease control was unsatisfactory and
a potential strategy might be to perform allogeneic transplant earlier
in the course of the disease. |
Carellaref |
16 with stage III MM |
51 |
|
|
|
|
|
|
9 patients are alive in remission at a median of 30 months after their
transplants, 1 patient is alive in relapse and 6 patients died of progressive
disease (5) or extensive chronic GvHD, infections and progressive disease
(1). |
In both studies, all patients had sustained allogeneic engraftment, and
at 100 days transplant-related mortality was low. However, due to acute
or or chronic GVHD complications, 1-year nonrelapse mortality was 15% and
11%, respectively. Allogeneic transplantation with reduced-intensity regimens
can be safety proposed to patients older than 50 years. This approach is
highly effective, with a CR rate over 50%. Progression free survival and
OS rates are encouraging, but the follow-up time is still short. Compared
to allogeneic transplantation with a myeloablative conditioning regimen,
miniallotransplantation is obviously associated with a much lower transplant-related
mortality. The incidence of acute GVHD remains relatively high and is probably
influenced by the conditioning regimen itself. Transplantation from unrelated
donors is feasibleref
and could yield results comparable to those achieved with HLA-identical
siblings. Conditioning regimens with high doses of antithymocyte globulin
or with Campath 1-H could reduce the incidence of severe acute GVHD. The
optimal procedure (conditioning regimen, type and duration of posttransplant
immunosuppression, interval between auto- and allogeneic HSCT, and the
role of DLIs) remains to be determined. Longer follow-up is needed to evaluate
the impact of this strategy on EFS and OS. Prospective multicenter trials
comparing tandem autologous HSCT and tandem autologous/allogeneic HSCT
are ongoing in the US and in Europe. Preliminary results of the IFM 9903-04
trial were presented at the 2003 ASH meetingref.
This trial recruited patients with newly diagnosed MM up to the age of
65 and with 2 adverse prognostic factors (ß2M level >
3 mg/L, presence of chromosome 13 deletion by FISH analysis). After four
courses of VAD (vincristine, doxorubicin, dexamethasone), these patients
received Mel 200 with ASCT. Then, according to the availibility of an HLA-identical
sibling, they received either an autograft with reduced-intensity conditoning
or a second allograft prepared by Mel 220 mg/m2 (± anti-IL-6
antibody). There was no significant difference between the two strategies
in terms of EFS or OS (both on an ITT analysis and when comparing actually
received treatments). Although the follow-up is still short (median follow-up
time 18 months), these results can be interpreted since the outcome in
this subgroup of patients is usually poor even with tandem ASCT. Moreover
it has been recently shown that deletion 13q detected by FISH remains a
negative prognostic factor after allograft with reduced-intensity conditioningref.
In this retrospective multicenter study, patients with 13q deletion (n
= 31) had a lower 2-year EFS rate (18% vs 42%) and a lower 2-year OS rate
(18% vs 47%). The major question regarding the use of allotransplantation
in patients with newly diagnosed MM remains: Should it be proposed to all
patients with an HLA-identical sibling (or even an HLA-identical unrelated
donor) or should it be proposed to selected patients? For instance, considering
the very good results obtained with tandem ASCT in patients with no adverse
prognostic factors, is it justified to propose a therapy that carries a
high risk of chronic GVHD and a 10%-15% incidence of toxic death? This
question remains to be answered by ongoing prospective multicenter trials.
Different g/d T clones sustain GVM (MRD
eradication) and aGVHD effects in MM patients after non-myeloablative transplantationref.
Sustained molecular remission by RIC allogeneic HSCT after autologous
HSCT in a patient with MMref.
Associated with a high incidence of extramedullary relapsesref.
The combination of high-dose therapy and autologous HSCT followed by
RIC HSCT was investigated to extend the benefit of allografting procedures
in de novo MM patients. 15 subjects with stage III MM (median age
51 years, range 40-57) received high dose melphalan (200 mg/m2)
followed by APBSCT previously collected after cyclophosphamide (4 g/m2)
and G-CSF. After 3-4 months from APBSCT, the patients underwent RICT, consisting
of fludarabine 30 mg/m2 + cyclophosphamide 300 mg/m2
on days -4, -3, and -2. AGVHD occurred in 2 patients; 6 patients developed
chronic GVHD; 4 patients developed CMV antigenemia and were treated pre-emptively
with ganciclovir. No transplant related mortality was shown. Response was
simultaneously measured by both electrophoresis (EP) and immunofixation
(IF); when IF was negative, patients were classified in CR and when it
remained positive, nCR. After a median follow up of 44 months post APBSCT,
100 and 43% of patients are still alive and progression-free, respectively.
Overall, the CR + nCR rate after dose-reduced allograft was enhanced from
26.7 to 73.3%. A correlation not statistically significant between GVHD
and remission was foundref.
Among patients with newly diagnosed myeloma, survival in recipients
of a autologous HSCT followed by a stem-cell allograft from an HLA-identical
sibling is superior to that in recipients of tandem stem-cell autografts.
A trial enrolled 162 consecutive patients with newly diagnosed myeloma
who were 65 years of age or younger and who had at least one sibling. All
patients were initially treated with VAD, followed by melphalan and autologous
HSCT. Patients with an HLA-identical sibling then received nonmyeloablative
TBI (2 Gy) and stem cells from the sibling. Patients without an HLA-identical
sibling received 2 consecutive myeloablative doses of melphalan (intermediate
doses (100 mg/m2) or high doses (140 to 200 mg/m2)),
each of which was followed by autologous stem-cell rescue. The primary
end points were OS and EFS. After a median follow-up of 45 months (range,
21 to 90), the median overall survival and event-free survival were longer
in the 80 patients with HLA-identical siblings than in the 82 patients
without HLA-identical siblings (80 months vs. 54 months, P=0.01; and 35
months vs. 29 months, P=0.02, respectively). Among patients who completed
their assigned treatment protocols, treatment-related mortality did not
differ significantly between the double-autologous HSCT group (46 patients)
and the autograft–allograft group (58 patients, P=0.09), but disease-related
mortality was significantly higher in the double-autologous HSCT group
(43% vs. 7%, P<0.001). The cumulative incidence rates of grades II,
III, and IV GVHD combined and of grade IV GVHD in the autograft–allograft
group were 43% and 4%, respectively. Overall, 21 of 58 patients (36%) were
in complete remission after a median follow-up of 38 months (range, 10
to 72) after allografting. Of the 46 patients who received two autografts,
25 (54%) diedref.
Comments
: 46 patients with no siblings were excluded from the primary analysis
and did not receive protocol therapy. Although the results in the autograft–allograft
group were promising, the results in the double-autologous HSCT group were
unexpectedly poor. The feasibility of tandem autologous HSCT was only 56%,
since 46 of 82 patients without HLA-identical siblings completed the double-autologous
HSCT program. This feasibility rate is one of the lowest ever reported,
and recent large trials of double autologous HSCT yielded feasibility rates
of 70 to 80%ref1,
ref2.
Moreover, the median OSl was 58 months among these 46 patients, whereas
in an ITT analysis performed in the last Intergroupe Francophone du Myélome
trial (IFM99) involving protocols of double autologous HSCT, the projected
5-year OS rate was 62% among 889 patients (Harousseau JL, Attal M, Moreau
P, Garban F, Facon T, Avet-Loiseau H. The prognostic impact of CR + VGPR
in a double-transplantation program for newly diagnosed multiple myeloma
(MM): combined result of the IFM99 trials. Blood 2006;108:877a-877a). This
finding raises a question about patient selection, especially
since the percentage of patients with an HLA-identical sibling was strikingly
high (49%) and since complete cytogenetic data at diagnosis were not available
in this trial. Of the 59 patients enrolled in Bruno's study who were treated
according to the high-dose, melphalan-based, double-autologous-transplant
protocol, as compared with those treated according to the autograft–allograft
protocol, 9 (15%) received melphalan at a dose of 140 mg/m2
and 50 (85%) received a dose of 200 mg/m2. In these 2 groups,
7 of the 9 patients (78%) and 39 of the 50 patients (78%), respectively,
received 2 transplants. As reported, 20 other patients received melphalan
at a dose of 100 mg/m2, and 17 of these patients (85%) underwent
two transplantations. As suggested by Rajkumar and Kyle, a comparison of
survival among the 80 patients who had an HLA-identical sibling with survival
among the 128 patients who did not have an HLA-identical sibling or any
sibling still showed a significant advantage for those patients who had
an HLA-identical sibling (hazard ratio for death, 0.56; 95% confidence
interval [CI], 0.34 to 0.93; P=0.02; hazard ratio for an event, 0.66; 95%
CI, 0.46 to 0.94; P=0.02). In response to Moreau and colleagues, 46 of
the 59 patients enrolled in the high-dose, melphalan-based, double-autologous-transplant
protocol completed their assigned treatment, with a feasibility rate of
78%. Attal et al. previously reported median OS and EFS of 58 and 30 months,
respectively, after double autologous transplantationref,
a finding that is consistent with the results we report. No cytogenetic
information was given. According to Mendel's inheritance laws, there is
a 25% probability that two siblings are HLA-identical. Thus, the genetic
chance of having a family donor depends on several variables, including
family size and sibling age. It is estimated that some 35% of white patients
may have an HLA-identical sibling who is eligible to be a donor. Of all
199 patients with siblings in our study, 75 (38%) had such a siblingref
Combined kidney
and nonmyeloablative marrow transplantation
: 6 patients with renal failure due to MM received simultaneous kidney
and HSCT from HLA-identical sibling donors following nonmyeloablative conditioning,
including cyclophosphamide (CP), peritransplant antithymocyte globulin
and thymic irradiation. Cyclosporine (CyA) was given for approximately
2 months posttransplant, followed by donor leukocyte infusions. All 6 patients
accepted their kidney grafts long-term. Three patients lost detectable
chimerism but accepted their kidney grafts off immunosuppression for 1.3
to >7 years. 1 such patient had strong antidonor CTL responses in association
with marrow rejection. 2 patients achieved full donor chimerism, but resumed
immunosuppression to treat GvHD. Only 1 patient experienced rejection following
CyA withdrawal. He responded to immunosuppression, which was later successfully
withdrawn. The rejection episode was associated with antidonor Th
reactivity. Patients showed CTL unresponsiveness to cultured donor renal
tubular epithelial cells. Initially recovering T cells were memory cells
and were enriched for CD4+CD25+ cells. Three patients
are in sustained complete remissions of MM, despite loss of chimerism in
2. Combined kidney/BMT with nonmyeloablative conditioning can achieve renal
allograft tolerance and excellent myeloma responses, even in the presence
of donor marrow rejection and antidonor alloresponses in vitroref.
For patients who relapse after allogeneic HSCT :
-
bortezomib
: in 23 patients bortezomib was given as single agent to 9 patients (39%)
and in combination with steroids in the other 14 (61%). Major toxicities
were thrombocytopenia (10/23, 43%) and peripheral neuropathy (12/23, 52%).
The overall response rate was 61% (14/23), including 22% (5/23) immunofixation-negative
complete remissions. No significant differences in toxicity and response
rates were seen between patients treated with bortezomib plus steroids
and bortezomib alone. After a median follow-up of 6 months, PFS was 6 months.
21 patients are alive, 2 and 5 in continuous VGPR and CR, respectivelyref.
-
arsenic trioxide (ATO)
: a heavily pretreated patient with GvHD after HSCTs with poor clinical
status tolerated the treatment very well and had a remarkable clinical
response and achieved CRref
-
immunotherapyref
: g-rays doses of 10,000 and 15,000 cGy were
not sufficient to totally block cell replication in both cell lines and
primary tumors; cell replication was able to be inhibited completely only
at 18,000 cGy. Lower doses (10,000 cGy) and lethal doses of irradiation
(i.e., 15,000 and 18,000 cGy) dose-dependently increased the expression
of all surface antigens present on the cells before irradiationref.
Using SEREX on cell line HMy2, ring finger protein 167, KLF10, TPT1, p02
protein, cDNA FLJ46859 fis, and DNMT1, were related to the development
of different tumors. Bioinformatics was performed to predict 12 novel MMSA
(multiple myeloma special antigen) genes. The prediction of tumor antigens
provides potential targets for the immunotherapy of patients with MM and
help in the understanding of carcinogenesis. Crude lysate ELISA methodology
indicated that the optical density value of MMSA-3 and MMSA-7 were significantly
higher in MM patients than in healthy donors. Furthermore, SYBR Green real-time
PCR showed that MMSA-1 presented with a high number of copy messages in
MM. In summary, the antigens identified in this study may be potential
candidates for diagnosis and targets for immunotherapy in MMref
-
vaccines :
-
TSAs : idiotype
-
idiotypic or DNA vaccination

-
subcutaneous (s.c.) injections of autologous Id, conjugated to keyhole
limpet hemocyanin (KLH) and in association with low doses of GM-CSF induced
anti-KLH IgM and IgG (90% of patients), anti-KLH IgE (30%), anti-GM-CSF
IgG (20%), anti-Id IgG (20%), and anti-Id IgE (30%) for almost 2 years
after the end of treatment. Id-specific delayed type hypersensitivity skin
tests were positive in 85% of tested patientsref
-
idiotype pulsed dendritic
cells cancer vaccine
(Mylovenge©
)
appears now to be a feasible way to enhance innate or acquired immunity
to help eliminate minimal residual disease (MRD) following autologous
peripheral blood stem cell transplantation (ASCT)
-
TAA : known genes, including ring
finger protein 167, KLF10,
TPT1
/ p02 protein, cDNA FLJ46859 fis, and DNMT1
,
were related to the development of different tumors. Bioinformatics was
performed to predict 12 novel multiple myeloma special antigen (MMSA)
genesref.
The expression of 11 cancer/testis
antigens (CTAs)
was analyzed in bone marrow samples from MM patients (N=55) and healthy
donors (N=32) using RT-PCR. CTAs were frequently expressed in MM with 56%
(MAGEC2), 55% (MAGEA3), 35% (SSX1), 20% (SSX4, SSX5), 16% (SSX2), 15% (BAGE),
7% (NY-ESO-1), and 6% (ADAM2, LIPI) expressing the given antigen. Importantly,
CT antigens were not expressed in healthy bone marrow. Analyzing MM patients
(N=66) for antibody responses against MAGEA3, SSX2, and NY-ESO-1, strong
antibody responses were found against CTAs preferentially in patients who
had received allogeneic stem cell transplantation (alloSCT). Antibody responses
against NY-ESO-1 correlated with NY-ESO-1-specific CD4+ and
CD8+ T cell responses against peptide NY-ESO-151-62 and CD4+
responses against NY-ESO-1121-140 in one of these patients. These allogeneic
immune responses were not detectable in pre-transplant samples and in the
patients' stem cell donors indicating that CTAs might indeed represent
natural targets for graft-versus-myeloma effects. Immune responses induced
by alloSCT could be boosted by active CTA-specific immunotherapy which
might help to achieve long-lasting remissions in patients with MMref.
NY-ESO-1 antigen is expressed in >60% of poor-prognosis myeloma at diagnosisref1,
ref2.
-
monoclonal antibodies : there are relatively few surface antigens
on the plasma cells that are suitable for antibody-directed treatment.
Possible molecules includeref
...
-
immuno-gene therapy

-
plasma exchange

-
bisphosphonates
to diminish the painful skeletal complications
-
combined kidney
transplantation
and HSCT
in patients with end-stage
renal disease (ESRD)
to achieve donor-specific allotolerance through the induction of mixed
lymphohematopoietic chimerism.
-
conditionally
replicating measles virus

Response criteria
in MMref1,
ref2
:
-
complete remission (CR) : absence of paraprotein on serum and urine
electrophoresis (negative immunofixation) and < 5% plasma cells in the
marrow
-
very good partial response (VGPR) : decrease of 90% in the serum
paraprotein level
-
partial remission (PR) : decrease of 50% in the serum paraprotein
level and/or a decrease of 90% in the urine BJ protein
-
minimal response : decrease of 25% in the serum paraprotein level.
-
Summary of similarities and specific changes introduced in the New Uniform
Response Criteria compared to the EBMT/IBMTR Criteria
-
for patients with measurable levels of serum and urine monoclonal protein
levels, the criteria for CR, PR and progressive disease remain unchanged
-
clarification and revision of important practical details of response evaluation
-
elimination of mandatory 6 weeks wait time to confirm achievement of response
-
introduction of a similar non-time-dependent confirmation for relapse and/or
disease progression
-
clarification of the 'start time' for duration of response evaluation
-
requirement of PR as response requirement for new drug trials
-
allow use of quantitative immunoglobulin levels in patients in whom the
M-protein measurements are unavailable or unreliable
-
introduction of new response categories
-
elimination of the minor response category
-
incorporation of response criteria for the serum FLC assay to enable assessment
of response in patients with non- or oligo-secretory disease (Tables 5
and 6)
-
clarification that criteria for progressive disease (rather than criteria
for 'relapse from CR') are to be used for calculation of time to progression
and progression-free survival in patients who are in CR. Criteria for relapse
from CR are to be used only if DFS is calculated and reported
-
introduction of new category of clinical relapse or progressive disease
(Table 6)
-
introduces clinical relapse as a new optional end point
Prognosis : average
life expectancy : 3.5 yearsref1,ref2.
Trends in 5 year survival rates (%) of MM (USA)
-
1950-54 : 6%
-
1960-63 : 12%
-
1970-73 : 19%
-
1974-76 : 24.1%
-
1977-79 : 26.3%
-
1980-82 : 28.1%
-
1983-85 : 28.0%
-
1986-88 : 29.3%
-
1989-91 : 29.6%
-
1992-97 : 28.7%
5 year survival rates (%), by age at diagnosis :
|
1989-1992
|
1989-1993
|
1989-1994
|
1989-1995
|
1989-1996
|
<45 |
47.4 |
44.5 |
41.7 |
44.6 |
46.8 |
45-54 |
36.2 |
37.8 |
41.4 |
40.7 |
41.8 |
55-64 |
33.8 |
33.7 |
32.8 |
34.2 |
33.2 |
65-75 |
26.6 |
27.7 |
26.4 |
25.6 |
25.9 |
> 75 |
19.5 |
19.7 |
20.4 |
23.6 |
18.8 |
A patient with MM was being maintained on IFN-a
after VAD and autologous HSCT (pretreated with melphalan). An episode of
immune thrombocytopenia and (Coombs positive) autoimmune hemolytic anemia
(AIHA) was noted while on maintenance INF-a,
which remitted when it was withdrawn. Following this event, he achieved
a state of stable disease that persists (> 3 years) with no specific MM
treatment. This sequence of events suggests a relationship between an immunological
reaction induced by IFN-a and the prolonged
phase of stable diseaseref
The presence of somatic hypermutations of the immunoglobulin variable
region genes in MM plasma cells suggests that malignant transformation
occurs in a B cell that has traversed the germinal centers of lymph nodes.
However, the hypoproliferative nature of myeloma has led to the hypothesis
that the bulk of the tumor arises from a transformed B cell with the capacity
for both self-renewal and production of terminally differentiated progenyref1,
ref2,
ref3.
The clinical course of patients requiring therapy for myeloma varies markedly.
Even with tandem autotransplants yielding complete remission (CR) rates
in excess of 60%, survival ranges from a few months to > 15 years. The
extended time (almost 2 years) for those patients to achieve CR, and the
even longer time to achieve MRI-CR, strongly suggests enormous tumor cell
population heterogeneity in terms of drug responsiveness/resistance.
-
traditional prognostic factors account for only 15-20% of outcome heterogeneity
:
-
[b2-microglobulin]serum
> 4 mg/L at diagnosis
-
high [CRP
]serum
-
serum albumin
-
abnormal metaphase karyotypes, present in 33% of newly diagnosed patients
and reflecting stroma independence, have been consistently associated with
a rapidly fatal outcome, and < 10% of patients with these abnormalities
survive > 5 years
-
primary translocations involving the immunoglobulin heavy chain locus at
14q32 in 40% of patientsref.
According to a consensus report of a recent Paris workshop on myeloma genetics,
hyperdiploid and t(11;14)(q13,q32)+ myeloma are associated with
a good prognosis, whereas non-hyperdiploidy, often associated with translocations
other than t(11;14) and chromosome 13 deletion, imparts a strikingly dismal
prognosisref.
-
gene expression profiling (GEP) has become a routine method for the detailed
analysis of gene activity in normal cell development and the disease process.
Comparative GEP has been used with a limited number of genes (~6800) from
CD138-enriched plasma cell RNA, obtained from patients with newly diagnosed
myeloma, patients with MGUS, and normal healthy subjects, to identify genes
that might play a role in the initiation and progression of myelomaref1,
ref2.7,8
These studies revealed that newly diagnosed myeloma could be classified
into four subgroups, with the two extremes being similar to either MGUS
or myeloma cell lines. The myelomas with a myeloma cell line-like GEP signature
also tended to have poor-risk features at presentation, such as elevated
serum ß2M (> 4 mg/mL) and cytogenetic abnormalities. These
studies also revealed that, whereas MGUS and myeloma could be distinguished
from normal plasma cells, these two conditions were difficult, if not impossible,
to differentiate from each other. It is curious that MGUS appears to harbor
all the hallmarks of overt malignancy, yet this condition rarely converts
to overt myeloma requiring systemic therapyref.
These data suggest that changes in the bone marrow microenvironment or
failure of immune surveillance, rather than a genetic change in the tumor
cell, may account for the malignant conversion of this benign plasma cell
dyscrasia. Although myeloma manifests itself at the terminal differentiated
stage of the B cell lineage, there is much speculation about the actual
origins of the disease. Thus, to place myeloma in the context of plasma
cell differentiation, we determined the molecular signatures of cells representing
the late stages of human B cell development and showed that the GEP-defined
high-risk form of myeloma has a tonsil B cell-like signature, whereas cells
with low-risk features tend to be either bone marrow or tonsil plasma cell-likeref.
GEP profiling of CD138-selected plasma cells and CD19-selected B cells
from the bone marrow of patients with Waldenström's macroglobulinemia
revealed that both compartments exhibit signs of malignancy, with the plasma
cells having either a tonsil plasma cell-like or a myeloma-like GEP signature
(Shaughnessy J, Zhan F, Tian E, et al. Gene expression profiling of CD19
and CD138-enriched cells from Waldenstrom's macroglobulinemia (WM) reveals
distinct classes and novel WM-specific genes [abstract]. Blood. 2002;100:1227abstract).
Taken together, these data support the concept that Waldenström's
macroglobulinemia, and possibly myeloma, may be derived from either terminally
differentiated cells with various degrees of plasticity or from cells transformed
at distinct stages of late-stage B-cell differentiation, with the ability
to self-renew and produce progeny with partially differentiated phenotypes
and transcriptome profiles. Nearly 40% of myelomas harbor 1 of 5 recurrent
chromosomal translocations involving the immunoglobulin heavy (IGH) chain
locusref.
Given the transcription-activating nature of IGH translocations, GEP represents
a valuable tool for identifying all myeloma-specific IGH translocationsref1,
ref2,
ref3.
Furthermore, genes that exhibit highly correlated expression patterns with
these activated oncogenes may point to downstream targets of the deregulated
genetic pathways resulting from their hyperactivation. Approximately 50%
of myelomas harbor deletion of chromosome 13, and this abnormality is linked
to poor survivalref.
We have mapped a putative myeloma tumor suppressor gene to a minimal region
of deletion at 13q14, and we have recently used a combination of triple-color-interphase
fluorescence in situ hybridization (FISH) and GEP to predict, with a high
degree of accuracy, chromosome 13 deletion by GEP aloneref
(Zhan F, Hardin J, Bumm KH, et al. Global GEP can be used to accurately
predict chromosome 13 deletion in MM [abstract]. Blood. 2001;98:1553).
Finally, we have used GEP to investigate the mechanisms by which myeloma
plasma cells contribute to the development of osteolytic bone lesions.
Comparative GEP between myeloma patients with and without bone lesions
demonstrated that elevated expression of the Wnt signaling inhibitor DKK1
by myeloma plasma cells is highly correlated with bone disease and that
treatment of osteoblast precursors with serum from myeloma patients with
high serum DKK1 could block BMP2/Wnt-induced differentiation of these cells
into osteoblasts, suggesting a mechanism by which myeloma cells directly
contribute to the development of osteolytic bone lesions in this diseaseref
-
gene expression profiling to model the effectiveness of tandem autologous
HSCT for the treatment of MM : GEP has been used to develop risk-adapted
prognostic models for leukemias and lymphomasref1,
ref2,
ref3,
ref4,
ref5,
ref6,
ref7.
The highly variable outcome in patients with MM, with very little of this
variability being accounted for by current laboratory tests, prompted us
to investigate if GEP could better model event-free (EFS) and overall (OS)
survival in this disease as well. The GEP studies to be described are on
CD138-enriched (> 90% CD38+/CD45-) plasma cells from
bone marrow aspirates taken from newly diagnosed myeloma patients entering
the National Cancer Institute-sponsored Phase III clinical trial (Total
Therapy II), which enrolled 660 patients from 1998 to 2003. In addition
to tandem transplants with melphalan (200 mg/m2), Total Therapy
II also randomized patients upfront to thalidomide or no thalidomide and
provided one year of intensive consolidation therapy and an additional
year of dexamethasone maintenance. Since 2000, we have performed GEP on
pretreatment plasma cells from 351 consecutive patients entering the Total
Therapy II trial with a median follow up of 25 months.
-
Unsupervised Hierarchical Clustering of Global Gene Expression Patterns
of Pretreatment Myeloma Plasma Cells Defines Biological and Clinical Subgroups
: unsupervised hierarchical cluster analysis, based on a set of 4580 highly
variable genes from ~10,000 tested (SD = 0.6) genes, segregated 221 myelomas
into four discrete subgroups. These groups were characterized by significant
differences in genetic features, such as chromosome ploidy (P < 0.01),
trisomy 11 (P < 0.001), and 14q32 IGH translocations (P < 0.01),
as well as by clinical parameters, such as IgA isotype, albumin (< 3.5
g/dL), ß2M (> 4 mg/L), creatinine (> 2 mg/d), MRI lesions (P <
0.05), and EFS (P = 0.002) and OS (P = 0.06). In the current unsupervised
hierarchical cluster analysis, we noted a nonrandom distribution of spiked
expression of recurrent translocation partners within the dendrogram-defined
subgroups (Figure 2; see Color Figures, page 511). The data revealed that
26 of 30 CCND1 spikes were clustered in one subgroup whereas 28 of 32 MMSET/FGFR3,
4 of 5 MAF, and 3 of 4 MAFB spikes were all located in the same cluster
branch. CCND1 expression, either low-level expression associated with chromosome
11 trisomy and hyperdiploid karyotypes or spiked expression linked to the
t(11;14)(p13;q32) translocation and presence of normal karyotypes, defined
two distinct subgroups with relatively low risk for relapse. Elevated expression
of proliferation-associated genes, whether in a hyperdiploid or hypodiploid
karyotype or MMSET/MAF/MAFB, defined 2 poor-prognosis subgroups. The MMSET/MAF/MAFB
groups was more frequently associated with hypodiploid karyotypes, an IgA
isotype, increased incidence of 14q32 translocations by FISH, and reduced
incidence of MRI lesions, whereas the CCND1 spike group was significantly
associated with normal cytogenetics. Taken together, our results highlight
the relationship between the transcriptome, cytogenetics, and the biological
and clinical features of myeloma. These findings extend and refine our
previous gene expression classification system and provide further evidence
that distinct molecular entities of myeloma exist.
Outcome in gene expression profiling (GEP)-defined myeloma subgroups.
Kaplan-Meier plots of overall survival (OS) (left panel) and event-free
survival (EFS) (right panel), dated from initiation of Total Therapy II,
according to the GEP subgroups. Numbers in brackets indicate 95% confidence
interval. Note that risk of relapse is significantly different between
the groups. As expected, Group IV has the shortest EFS, Group III exhibits
an intermediate risk, and Groups I and II, which are essentially the same,
have the best prognosis. Although the OS is not currently significant,
a trend similar to the EFS is evident. Groups I-IV are defined as above.
The cumulative number of patients does not equal 221 due to removal of
patients who had a diagnostic gene array but failed to start the protocol.
-
Cox Regression Modeling of Gene Expression Patterns in Pretreatment Plasma
Cells Identifies Three Genes Associated with Rapid Relapse : because microarrays
are not likely to become routine clinical tests, comprehensive global GEP
studies will probably be used to identify a small subset of genes whose
expression can be applied in the development of gene-based risk-adapted
patient stratification, as has been recently done in lymphomaref.
We used Cox regression modeling of gene expression on EFS in 212 newly
diagnosed myeloma patients using data from U95Av2 microarray (~10,000 genes).
The median follow-up at the time of this analysis was 20 months. There
were 34 events representing either disease-specific death or progression/relapse.
EFS was modeled by using standard prognostic values as well as by GEP using
the Affymetrix signal. A generalized estimate of R2 was obtained by using
the approach suggested by Cox and Snell (Cox DR, Snell EJ. The analysis
of binary data (ed Second). London: Chapman & Hall; 1989). Gene expression
values considered were based on significance of univariate association
with EFS. The median signal call was used as a cut point prior to modeling
inclusion. The 100 genes most significantly associated with EFS based on
the score test were potential variables in multivariate modeling. Using
standard prognostic variables only, the model that best fit disease-specific
EFS was FISH13 (chromosome 13 deletion detected by FISH) and the presence
of chromosomal abnormalities (CA).
Adjusting for CA and FISH13, three genes-RAN, ZHX-2, and CHC1L-were simultaneously
significant, each at the P < 0.005 level. After adjustment for other
model variables, patients with high RAN expression had increased risk of
event while patients with high ZHX-2 or high CHC1L had decreased risk of
event. After adjusting for these three genes and each other, FISH13 patients
had an increased risk of event but those with CA did not. R2 for the FISH13
and CA model was 30%. The R2 value for the RAN, ZHX-2, and CHC1L model
was 66%. It is important to note that each of these genes has independent
prognostic influence, so the combination of RAN overexpression and loss
of expression of ZXH-2 and CHC1L represents a much more powerful model
than any other combination
Association of folate transporter SLC19A1
polymorphisms with the outcome of multiple myeloma after chemotherapy and
tandem autologous transplantationref
High-risk myeloma defined by expression of 3 genes.
Cumulative incidence of event-free survival defined by the various
combinations of expression of 3 genes. Note that only high RAN/low ZHX-2/low
CHC1L shows dismal prognosis. It is important to note that the three-gene
combination accounts for 66% of patient outcome variability. The best current
models are near 30%.
We next compared the EFS between a high-risk entity (patients whose
plasma cells express RAN above the median and ZHX-2 and CHC1L below the
median; n = 45) and a low-risk entity (all remaining cases; n = 165). In
this population, there were 30 events in the 45 high-risk patients (66%)
and 30 events in the 165 low-risk patients (18%) (P < 0.0001). We next
calculated the EFS based on the combination of GEP risk groups and CA.
EFS rates were not significantly different between high-risk and low-risk
disease with and without CA. However, high-risk and low-risk disease groups
were significantly different irrespective of the presence of CA (P <
0.0001).
Gene expression profiling (GEP) model defines a high-risk disease and
is not affected by presence of abnormal cytogenetics.
Left panel presents Kaplan-Meier curves for high-risk myeloma (high
RAN/low ZHX-2/low CHC1L) and low-risk myeloma (all others). The right panel
shows Kaplan-Meier curves for high-risk and low-risk disease with respect
to the presence or absence of any cytogenetic abnormalities (CA). Note
that the model is not influenced by CA, implying that GEP high-risk myeloma
is a more robust indicator of poor prognosis than the current best prognostic
variable.
-
RAN, ZHX-2, and CHC1L Function Provides Insight into How These Genes Might
Impart a High-Risk Phenotype
-
RAN, which maps to 6p21, is a member of the Ras family of GTPase proteins
that has a role in many aspects of cell biology, including shuttling protein
and RNA in and out of the nucleus as well as regulating chromosome condensation,
spindle formation, nuclear assembly, and cell-cycle progressionref1,
ref2.
During interphase, RAN promotes nuclear localization signal-mediated protein
import through the association and dissociation of transport complexes.
A complex containing aster-promoting activities, importin /importin ß,
forms in the cytosol and translocates across the nuclear pore. In the nucleus,
Ran-GTP binds to importin ß and dissociates the transport complex.
Ran-GTP and importin ß shuttle back to the cytosol, where Ran-GTP
is hydrolyzed by cytosolic RanGAP1 and RanBP1, to form Ran-GDP. The polarized
distribution of Ran-GTP across the nuclear envelope is maintained by the
compartmentalization of RCC1 (RanGEF), RanGAP1, and RanBP1. The localization
of RCC1 on chromatin promotes microtubule assembly in mitosis. During mitosis,
importin and importin ß sequester APA in an inactive form in
regions where Ran-GTP is low. Increased levels of Ran-GTP near the chromosome
promote local disassembly of APA components from complexes containing importin
and importin ß. In high Ran-GTP regions, importin ß associates
with Ran-GTP, maintaining it in an inactive state until RanGAP1 and RanBP1
catalyze Ran-GTP hydrolysis. After dissociation from importin complexes,
APA becomes active in promoting microtubule assemblyref.
-
The CHC1L gene (CHromosome Condensation 1-Like, also called E4.5)ref
maps to 13q14.3 in a region suspected to harbor a tumor suppressor gene
for myelomaref1,
ref2,
ref3.
Although we have evidence that the chromosome 13 tumor suppressor gene
may be RB1ref,
this has not been proven. Reduced CHC1L expression has emerged as an integral
component of a three-gene model for high-risk disease, and, consequently,
it must be considered as a potential candidate tumor suppressor gene. Although
the function of the product of CHC1L has not been completely described,
it is a homologue of RCC1, which, as pointed out above, is the GTP exchange
factor (GEF) for RANref.
CHC1L has been proposed to be a candidate tumor suppressor gene in both
B-CLL30 and prostate cancerref.
Thus, a loss of function of CHC1L may somehow influence the function of
RAN and, together with the increased expression of RAN and loss of ZHX-2,
may impart the aggressive phenotype on myeloma with this particular gene
expression pattern. The poor MSK phenotype is conferred only by genetic
interaction, in that gain of RAN or loss of CHC1L alone does not confer
the aggressive phenotype. We are currently investigating whether CHC1L
may have a RanGEF function in myeloma, if myeloma with activated RAN and
loss of CHC1L has an altered ratio of nuclear and cytoplasmic RAN, and
whether levels of Ran-GTP in the nucleus are altered in cases or/altered
CHCTL.
-
ZHX-2, the third gene in the model, has recently been cloned and has been
shown to be a negative regulator of the NF-Y transcription factorref.
This has important implications as the NF-Y complex is a master transcriptional
regulator of many genes involved in cell-cycle control and proliferation,
including CCNB1, CCNB2, CCND2, CDC2, CDC25, TOP2A, TK1, and others. There
is a negative correlation between ZHX-2 expression and the expression of
a panel of 30 proliferation-associated genes, including genes known to
be positively regulated by NF-Y). The gene ZHX-2 maps to 8q24.3, very near
the MYC oncogene. The loss of chromosome 8 by cytogenetic analysis imparts
a very high risk of relapse, but not to death when patients are treated
with high-dose therapy and peripheral blood stem cell transplantsref.
Reduced expression of ZHX-2 was linked to poor survival in Total Therapy
II, which, based on its chromosomal locus, may provide a mechanistic explanation
for why loss of chromosome 8 by conventional cytogenetics imparted such
a high risk in Total Therapy Iref.
As would be expected from the results of the ZHX-2 correlation studies,
overexpression of proliferation-associated genes is also linked to poor
survival. Given the link between ZHX-2 loss and rapid development of resistance
in myeloma to combination chemotherapy, it is noteworthy that NF-Y-mediated
transactivation of the human ribonucleotide reductase subunit M2 is related
to Gemcitabine resistanceref.
The transcription factor homeobox B4 (HOXB4) gene is preferentially expressed
in immature hematopoietic cells and implicated in the transition from primitive
hematopoiesis to definitive hematopoiesis as well as in immature hematopoietic
cell proliferation and differentiation. Recent studies have shown that
NF-Y cooperates with USF1/2 to induce the hematopoietic expression of HOXB4ref.
Thus, it is possible that myeloma cells that lose ZXH-2 upregulate HOXB4,
and these cells acquire a stem cell-like phenotype and resistance to chemotherapy.
The mechanism by which ZXH-2 expression is reduced and its potential function
in multiple myeloma are currently under investigation. Elevated expression
of cell cycle- and proliferation-associated genes is associated with poor
event-free survival (EFS) (left) and overall survival (OS) (right).
Kaplan-Meier curves based on the 4 quartiles of CCNB1 (top), PCNA (middle),
and BUB1B (bottom) gene expression. Q1 is the lowest quartile and Q4 is
the highest. Note that all Q4 patients tend to do poorly.
Unsupervised hierarchical cluster analysis of gene expression patterns
in CD138-enriched plasma cells from untreated myeloma patients allowed
the identification of four subgroups of patients whose GEP signatures are
highly correlated with distinct cytogenetic abnormalities, clinical parameters,
and survival after high-dose therapy with peripheral blood stem cell transplant.
These data lend credence to the concept that multiple myeloma consists
of multiple disease entities with distinct mechanisms of transformation,
as evidenced by the highly coordinated gene expression changes seen in
each subtype. It is currently not clear whether this coordinated subgroup-specific
expression occurs through the deregulation of distinct molecular pathways.
However, if confirmed, these data could provide a framework for the development
of novel therapeutic interventions based on the molecular biology of disease
subtypes. Our experience over the last five years with the prospective
application of genomic analysis of tumor cells from a uniformly treated
population at diagnosis and follow-up suggests that myeloma is amenable
to genomic classification. We firmly believe that a molecular classification
of myeloma will prove useful in promoting the more effective application
of current therapies and provide the framework for the development of novel
therapeutic strategies based on common molecular features of each distinct
subtype.
Please note that data described here are preliminary and based on the
use of microarrays containing ~12,000 genes in a population of 221 patients.
These models represent the most current iterations in our attempts to create
a genomic classification of myeloma and have not been validated.
Applying Significance Analysis of Microarrays, 52 genes, involved in
important pathways related to cancer, were differentially expressed between
plasma cells from healthy subjects (n=22) and patients with stringently
defined MGUS/smoldering MM (n=24) and symptomatic MM (n=351) (P < .001).
Unsupervised hierarchical clustering of 351 MM, 44 MGUS (24 + 20) and 16
MM with MGUS history, created two major cluster branches, one containing
82% of the MGUS cases and 28% of the MM, termed MGUS-like MM (MGUS-L MM).
Using the same clustering approach on an independent cohort of 214 cases
of MM, 27% were MGUS-L. This molecular signature, despite being associated
with a lower incidence of complete remission (P = .006), was associated
with low-risk clinical and molecular features and superior survival (P
< .01). The MGUS-L signature was also seen in plasma cells from 15 of
20 patients surviving more than 10 years after autotransplantref.
Co-diagnoses :
Bibliography :
Web resources
:
Copyright © 2001-2014 Daniele Focosi.
All rights reserved
| Terms of use | Legal
notices
About
this site | Site map
| Acknowledgements | Current
link partners
Abbreviations
and acronyms | Medical
terminology | Add
a link | Translate
| Softwares
|
Cite
this page!